quiz image

Medical Microbiology: Bacteriology L2

NobleTucson avatar
NobleTucson
·
·
Download

Start Quiz

Study Flashcards

334 Questions

What is the significance of whole-genome sequencing in bacteriology?

It allows for the identification of virulence factors, antimicrobial resistance genes, and other genetic elements that contribute to the pathogenesis of bacterial infections.

What is the purpose of genome annotation?

To identify and describe the functional elements of a genome, such as genes, regulatory regions, and repetitive elements.

What are some methods used in analyzing a bacterial genome?

Gene prediction, functional annotation, comparative genomics, and phylogenetic analysis.

What are some applications of genome sequencing in bacteriology?

Outbreak investigation, vaccine development, antimicrobial resistance surveillance, and pathogen discovery.

What is the process of locating the position of genes in a genome called?

Genome Annotation

What is the purpose of read-mapping and de novo assembly in genome sequencing?

To map reads to a reference genome or assemble reads into a complete genome sequence

What is a coding DNA sequence (CDS) in genome annotation?

A protein-coding DNA sequence encoded for by a gene

What is the primary function of the BLAST computer program in genome annotation?

To perform base-by-base comparison of two or more gene sequences and assign tentative function of a gene or protein structure based on the alignment

How is the function of a gene annotated in a bacterium where the function has not been studied?

By using a chain of annotation, where the function is inferred from a similar gene sequence in another bacterium

What is the goal of bioinformatics in microbiology?

To address novel questions in microbiology, such as disease transmission, evolution, and pathogenesis, using genomic data and computational tools

What is the term for the study of genetic variation at a population level?

Population genetics

What is the purpose of mapping sequence reads to a reference genome?

To identify genetic variations, such as single nucleotide polymorphisms (SNPs), within a population

What is the term for a variation in a single nucleotide at a specific locus in a population?

Single nucleotide polymorphism (SNP)

What is the primary goal of phylogenetics in the context of genomic sequences?

To estimate the evolutionary relationships among a set of genome sequences

What is the foundation of other omic studies, such as transcriptomics and proteomics?

Genomics

What is the focus of genomic epidemiology?

Linking genomics with epidemiology

What is the role of selective pressure in the evolution of new traits?

It drives the evolution of new traits

What is the main application of phylogeography in disease transmission?

To infer how bacteria spread

What is the significance of genome sequencing in tracking disease transmission?

It provides the highest possible resolution for tracking disease transmission

What is the primary focus of epidemiology?

The occurrence, determinants, distribution, and control of health and disease in a defined human population

What is the relationship between phylogenetics and phylogeography?

Phylogeography combines phylogenetics with geographical mapping

What is the main difference between the Sec Translocase pathway and the Twin Arginine Transport (TAT) pathway in terms of protein secretion?

The Sec Translocase pathway secretes unfolded proteins, while the TAT pathway secretes folded proteins.

What is the purpose of a signal tag in the Sec Translocase pathway?

To target proteins for the Sec system.

What is the role of TatB/C in the TAT pathway?

Recognition of the twin arginine tag at the N-termini of proteins.

Why is it advantageous to transport folded proteins?

To prevent protein misfolding or aggregation during transport.

What is the name of the pore formed during the TAT pathway?

The TatA pore.

Can Sec translocation be a co-translational or post-translational process?

Yes.

What is the difference between the Sec and TAT pathways in terms of protein recognition?

The Sec pathway recognizes a signal tag, while the TAT pathway recognizes a twin arginine tag.

What is the role of the TatA ring in the TAT pathway?

It allows the Tat protein to translocate through.

What is the primary function of the Dot/Icm system in Legionella pneumophila and Coxiella burnetii?

Translocation of proteins from the cytoplasm and periplasm to block, blunt, and subvert host cells

What is the mechanism of secretion used by the T6SS system?

Contact-dependent secretion mechanism

What is the estimated percentage of Gram-negative bacteria that possess a T6SS system?

25%

What is the term 'Icm' an abbreviation for in the context of the Dot/Icm system?

Intracellular multiplication

What is the term 'Dot' an abbreviation for in the context of the Dot/Icm system?

Defective organelle trafficking

Which bacterium is a prototypic example of a species that possesses a T6SS system?

Pseudomonas aeruginosa

What is the name of the mechanism by which T6SS can be used to target other bacteria?

Inter-bacterial combat

How many T6SS systems are found in the bacterium Burkholderia thailandensis?

Five

What is the primary function of the T5SS system in Gram-negative bacteria?

The primary function of the T5SS system is to allow the translocation of proteins across the plasma membrane after Sec translocation.

What are the four key elements of T5SS proteins?

The four key elements of T5SS proteins are an N-terminal signal tag, an internal passenger domain, a linker region, and a C-terminal β barrel/translocator domain.

What is the role of the BAM complex in T5SS?

The BAM complex mediates the insertion of the translocator domain of T5SS proteins into the outer membrane.

What is the difference between T5SS anchored and secreted proteins?

T5SS anchored proteins remain attached to the surface of the bacterium, while T5SS secreted proteins are released into the environment after cleavage.

What is the significance of T1SS and T5SS in bacterial virulence?

T1SS and T5SS proteins can be associated with virulence, as they enable the secretion of proteins that can subvert the host response and contribute to the pathogenesis of bacterial infections.

What is the purpose of dedicated secretion systems in Gram-negative pathogens?

Dedicated secretion systems, such as T5SS, enable the translocation of proteins across three membranes (inner, outer, and host) and are essential for the survival of Gram-negative pathogens.

What are Effectors in the context of bacterial secretion systems?

Effectors are proteins that are secreted through dedicated secretion systems, such as T5SS, and are translocated into host cells, where they can subvert the host response and contribute to the pathogenesis of bacterial infections.

What is the significance of secretion systems in the pathogenesis of bacterial infections?

Secretion systems, such as T5SS, play a crucial role in the pathogenesis of bacterial infections by enabling the secretion of proteins that can subvert the host response and contribute to the virulence of Gram-negative pathogens.

What is the function of the cognate immunity proteins in the Type 6 Secretion System (T6SS)?

These proteins protect the bacterial cell from the toxins injected by the T6SS.

What is unique about the Type 7 Secretion System (T7SS) in Gram-positive bacteria?

It is capable of transporting proteins across the inner membrane and the mycobacterial cell wall.

What is the role of the C-terminal signal tag in the Type 7 Secretion System (T7SS)?

It is required for protein transport across the inner membrane and the mycobacterial cell wall.

Why is the Type 6 Secretion System (T6SS) used in bacterial competition?

It allows bacteria to inject toxins into neighboring cells, giving them a competitive advantage.

What is a characteristic of the Type 7 Secretion System (T7SS) in Mycobacterium tuberculosis?

It requires folded proteins for transport.

What is the significance of the Type 7 Secretion System (T7SS) in Mycobacterium tuberculosis?

It is required for virulence.

How do bacteria with a Type 6 Secretion System (T6SS) protect themselves from their own toxins?

They produce cognate immunity proteins that block the effect of the toxins.

What is the 'tit for tat' phenomenon in the context of the Type 6 Secretion System (T6SS)?

It is a form of bacterial retaliation, where cells respond to toxin injection by producing their own toxins.

What is the main function of T4SS in bacteria, and what type of molecules do they translocate?

T4SS enable the translocation of DNA or proteins to other bacteria as well as eukaryotic cells

What are the two main subfamilies of T4SS, and what do they translocate?

Conjugation machineries (DNA and protein transfer) and effector protein translocation systems (protein transfer)

What is the prototypic VirB/VirD system, and what is its function?

The prototypic VirB/VirD system is found in Agrobacterium tumefaciens and delivers a fragment of the A. tumefaciens genome to the host cell, leading to Crown Gall Disease in plants

What is the mechanism of DNA/protein transfer in the VirB/VirD system?

DNA/protein transfer occurs in a single step process from the cytoplasm to outside the cell, via a pilus (VirB2) enabling cell-to-cell transfer

What is the role of the pilus (VirB2) in the VirB/VirD system?

The pilus (VirB2) enables cell-to-cell transfer between the bacterial and target cell

What type of disease is caused by the Agrobacterium tumefaciens VirB/VirD system?

Crown Gall Disease in plants

What is the relationship between the VirB/VirD system and T4SS?

The VirB/VirD system is a type of T4SS, specifically a conjugation machinery

What are some examples of bacteria that use the VirB/VirD system?

Helicobacter pylori Cag system and Bordetella pertussis pertussis toxin secretion

Which secretion system translocates proteins into host cells using an N-terminus secretion signal?

T3SS

What is the primary function of the T5SS system in Gram-negative bacteria?

Autotransporter pathway

Which secretion system is used by Legionella and Coxiella to transfer proteins?

T4SS

What is the significance of the T6SS system in Gram-negative bacteria?

Translocates proteins into host cells and target other bacteria

What is the primary function of the Dot/Icm system in Legionella pneumophila and Coxiella burnetii?

Type IV secretion system

What is the role of the BAM complex in T5SS?

Outer membrane assembly

What is the significance of secretion systems in the pathogenesis of bacterial infections?

Transfer of virulence factors into host cells

What is the primary function of dedicated secretion systems in Gram-negative pathogens?

Transfer of virulence factors into host cells

How do Sec and Tat secreted proteins contribute to virulence in bacteria?

Sec and Tat secreted proteins contribute to virulence by contributing to host colonization and remodeling peptidoglycan. For example, autolysins of Listeria monocytogenes contribute to host colonization by remodeling peptidoglycan, and Phospholipase C of Pseudomonas aeruginosa requires Tat-based secretion to be functional.

What is a key difference between Gram-negative and Gram-positive bacteria in terms of protein secretion?

Gram-negative bacteria have two membranes (inner and outer membranes), requiring unique mechanisms for protein secretion, whereas Gram-positive bacteria have a single membrane.

What is the significance of the outer membrane in Gram-negative bacteria in the context of protein secretion?

The outer membrane requires specialized transport systems to allow proteins to traverse it, which are not needed in Gram-positive bacteria.

How do Type I secretion systems (T1SS) differ from other secretion systems in Gram-negative bacteria?

T1SS are unique in that they can secrete proteins directly from the cytoplasm to the extracellular environment in a single step.

What is the advantage of secreting folded proteins via the Tat pathway?

The Tat pathway allows for the secretion of folded proteins, which can be advantageous for proteins that require specific conformations for function.

What is the role of the TatA ring in the TAT pathway?

The TatA ring forms a pore in the membrane that allows for the secretion of folded proteins.

How do secretion systems contribute to bacterial virulence?

Secretion systems allow bacteria to secrete proteins that contribute to host colonization, remodeling of peptidoglycan, and other processes that facilitate pathogenesis.

What is the significance of dedicated secretion systems in Gram-negative pathogens?

Dedicated secretion systems allow for the targeted secretion of specific proteins that contribute to virulence, enhancing the bacterium's ability to cause disease.

What is the characteristic lesion triggered by Enteropathogenic E. coli (EPEC)?

attaching and effacing lesion

What is the type of secretion system triggered by contact between a pathogen and a target eukaryotic cell?

contact-dependent Type III secretion

What is the function of chaperones in the Type III secretion system?

guide specific secreted proteins to the secretion apparatus

What is the name of the pathogenicity island found in E. coli E2348/69?

locus of enterocyte effacement (LEE)

How many Type III secretion systems are found in most Salmonella?

two (SPI1 & SPI2)

What is unique about the organization of genes encoding effectors in Type III secretion systems?

genes encoding effectors do not have to be genetically linked to the T3SS machinery

What are the four key elements that T5SS proteins contain, and what is their role in protein secretion?

The four key elements of T5SS proteins are an N-terminal signal tag, an internal passenger domain, a linker region, and a C'-terminal β barrel/translocator domain. These elements facilitate the translocation of proteins across the plasma membrane after Sec translocation in a multi-step mechanism.

What is the role of the BAM complex in T5SS, and how does it facilitate protein secretion?

The BAM complex mediates the insertion of the translocator domain into the outer membrane, allowing for protein secretion through T5SS.

What is the difference between T5SS anchored and secreted proteins, and how do they contribute to bacterial virulence?

T5SS anchored proteins are attached to the surface of the bacterium, while secreted proteins are released into the environment. Both types contribute to bacterial virulence by facilitating the translocation of proteins across the plasma membrane.

What is the significance of T1SS and T5SS in bacterial virulence, and how do they contribute to pathogenesis?

T1SS and T5SS are important secretion systems that enable bacteria to translocate proteins across the plasma membrane, contributing to bacterial virulence and pathogenesis.

What is the purpose of dedicated secretion systems in Gram-negative pathogens, and how do they facilitate protein secretion?

Dedicated secretion systems, such as T1SS and T5SS, allow Gram-negative pathogens to translocate proteins across three membranes (inner, outer, and host), facilitating protein secretion and contributing to pathogenesis.

What are Effectors in the context of bacterial secretion systems, and what is their role in bacterial virulence?

Effectors are secreted proteins that are translocated into host cells, facilitating bacterial virulence and contributing to pathogenesis.

What is the significance of secretion systems in the pathogenesis of bacterial infections, and how do they contribute to disease?

Secretion systems, such as T1SS and T5SS, enable bacteria to translocate proteins across the plasma membrane, contributing to bacterial virulence and disease.

What is the main difference between T1SS and T5SS, and how do they contribute to bacterial virulence?

T1SS and T5SS are both secretion systems, but they differ in their mechanisms of protein translocation. T1SS uses a single protein to form a channel, while T5SS uses a β-barrel translocator domain. Both systems contribute to bacterial virulence by facilitating protein secretion.

What is the primary function of the VirB/VirD system in Agrobacterium tumefaciens?

To deliver a fragment of the A.tumefaciens genome to the host cell, known as oncogenic T-DNA, plus effector proteins, leading to Crown Gall Disease in plants.

What is the main difference between the conjugation machineries and effector protein translocation systems of T4SS?

Conjugation machineries transfer DNA and proteins via the VirB/VirD system, while effector protein translocation systems transfer proteins via the Dot/Icm system.

What is the role of the pilus (VirB2) in the VirB/VirD system?

The pilus (VirB2) enables cell-to-cell transfer between the bacterial and target cell, facilitating the delivery of DNA and proteins.

What is the mechanism of DNA/protein transfer in the VirB/VirD system?

DNA/protein transfer occurs in a single step process from the cytoplasm to outside the cell.

What is the significance of the Dot/Icm system in Legionella pneumophila and Coxiella burnetii?

The Dot/Icm system is a type of T4SS that enables the translocation of effectors proteins into host cells, contributing to the pathogenesis of Legionella pneumophila and Coxiella burnetii.

What is the primary function of T4SS in bacteria?

To enable the translocation of DNA or proteins to other bacteria as well as eukaryotic cells, facilitating conjugation, protein transfer, and pathogenesis.

What is the relationship between T4SS and conjugation systems in bacteria?

T4SS are evolutionarily related to bacterial conjugation systems, and share similarities in their mechanism of DNA and protein transfer.

What is the significance of T4SS in the pathogenesis of bacterial infections?

T4SS enable the translocation of effectors proteins and DNA into host cells, contributing to the pathogenesis of bacterial infections and disease development.

What is considered a novel secretion system used by all bacteria?

Bacterial membrane vesicles (MVs)

What is the composition of bacterial membrane vesicles (MVs)?

MVs contain lipopolysaccharides, proteins, DNA, and other bacterial components

How are bacterial membrane vesicles (MVs) generated?

MVs are generated through the budding of the outer membrane of Gram-negative bacteria and the fragmentation of the cell wall of Gram-positive bacteria

What is the role of environmental factors in bacterial membrane vesicle (MV) production?

Environmental factors can increase BMV production

What is the role of bacterial membrane vesicles (MVs) in mediating inflammation in the host?

BMVs produced by both Gram-negative and Gram-positive bacteria can mediate inflammation in the host by activating host innate immune receptors (PRRs)

What is the non-pathogenic role of bacterial membrane vesicles (MVs)?

BMVs can confer a selective advantage to bacteria, promoting bacterial survival

What is the difference between MVs produced by Gram-positive and Gram-negative bacteria?

MVs produced by Gram-positive and Gram-negative bacteria have different membrane structures and cargo

What is the role of bacterial membrane vesicles (MVs) in delivering virulence determinants?

BMVs can deliver virulence determinants to and into host cells, promoting pathogenesis

What is the significance of bacterial membrane vesicles (MVs) in bacterial communication?

MVs play a crucial role in bacterial communication, promoting bacterial adaptation and survival

What is the role of bacterial membrane vesicles (MVs) in promoting bacterial survival?

BMVs can promote bacterial survival by conferring a selective advantage to bacteria

What is the sensitivity of TB commonly diagnosed by staining?

36.8%

What is the mechanism used in Immunofluorescence to detect pathogens?

Fluorescent antibody binds directly to the pathogen and viewed under a microscope

What is the role of GeneXpert in TB diagnosis?

Automates DNA extraction and DNA amplification from sputum samples

How was Legionella pneumophila discovered?

By infected guinea pigs with lung tissue from infected patients and then infecting eggs with guinea pig tissue

What is the primary importance of diagnostic tests?

Combined understanding of the disease and correct interpretation of results

What are the two main categories of diagnostic tests?

Culture dependent and culture independent

What is the significance of molecular detection methods?

Used where organisms are difficult to culture or speed of diagnosis is essential

What is the name of the disease caused by Legionella pneumophila?

Legionnaires' disease

What is the role of PCR/real-time PCR in molecular detection?

Nucleic acid amplification

What is the advantage of automated systems in molecular detection?

Speed and ease of use

What is the primary difference between a pooled screen and other methods of CRISPR application?

Pooled screen uses next-gen sequencing to compare the abundance of different transgenes in each sample, while other methods may use individual wells or different readout methods.

How does CRISPR-Cas9 modify T-cells in cancer treatment?

CRISPR-Cas9 disables PD-1, which down-regulates the immune response, allowing T-cells to attack and defeat cancerous cells.

What is the significance of Anti-CRISPR mechanisms?

Anti-CRISPR mechanisms, such as mutations leading to mismatches and inefficient cleavage, can inhibit CRISPR-Cas activity.

What is the goal of genome editing in human embryos?

The goal is to correct genetic mutations or defects that can cause disease or disorders.

What is the significance of Transthyretin amyloidosis in the context of CRISPR?

Transthyretin amyloidosis is a heart disease that can be targeted by CRISPR-Cas9 gene editing.

What is the purpose of the podcast 'RadioLab episode: Antibodies Part 1: CRISPR'?

The podcast provides an educational and informative discussion about CRISPR technology and its applications.

How does the CRISPR-Cas system defend against bacteriophages?

CRISPR-Cas systems can defend against bacteriophages by incorporating phage-derived spacers into their CRISPR arrays, allowing them to recognize and target phage DNA.

What is the role of Cas enzyme in the CRISPR-Cas system?

The Cas enzyme is responsible for cleaving target DNA sequences that are complementary to the CRISPR RNA.

What is the significance of PAM sequences in the CRISPR-Cas system?

PAM sequences are necessary for the recognition and binding of the Cas enzyme to target DNA sequences.

What is the main advantage of using CRISPR-Cas9 for gene editing compared to other gene editing tools?

CRISPR-Cas9 is a precise and efficient method for gene editing, allowing for specific targeting of genetic sequences.

What is the primary adhesin used by Enterohaemorrhagic E. coli (EHEC)?

LEE, Paa, ToxB, Efa-1/LifA, LPF

What is the primary site of infection for Enteropathogenic E. coli (EPEC)?

Small bowel enterocytes

What is the primary adhesin used by Uropathogenic E. coli (UPEC)?

Fim, Pap, F1C, Dr

What is the primary site of infection for Meningitis-associated E. coli (MNEC)?

Meninges

What is the primary adhesin used by Avian pathogenic E. coli (APEC)?

Fim

What is the primary difference between Enterotoxigenic E. coli (ETEC) and Enteroaggregative E. coli (EAEC)?

Adhesins used, ETEC uses Fim and TCP, while EAEC uses Fim, AAF, and dispersin

What is the primary site of infection for Enteroinvasive E. coli (EIEC)?

Colon

What is the primary adhesin used by Diffusely adhering E. coli (DAEC)?

Fim, Dr adhesins

What is the primary difference between EHEC and EPEC?

Site of infection, EHEC causes HUS and EPEC causes diarrhea

What is the primary role of Fim in E. coli?

Adhesion to host cells

What is the primary mechanism by which cytosolic pathogens, such as Listeria and Shigella, spread within organs/tissues?

Actin tails promote bacterial spread within organs/tissues by driving bacteria into protrusions, which are then engulfed by neighbouring cells, allowing the bacteria to avoid host immunity.

How do intravacuolar pathogens, such as Salmonella, Mycobacteria, Chlamydia, and Legionella, protect themselves from cytoplasmic immune sensing molecules?

They establish intracellular niches by constructing membrane-encompassed compartments, which protect them from cytoplasmic immune sensing molecules.

What is the role of effectors in modulating host vesicle trafficking in Legionella pneumophila?

Effectors manipulate LCV surface molecules, making the LCV look like another ER, and prevent LCV-lysosome fusion, allowing the bacteria to survive.

What is the primary function of the Dot/Icm system in Legionella pneumophila and Coxiella burnetii?

The Dot/Icm system is a type IV secretion system that allows the bacteria to manipulate the host cell and establish a replicative niche.

How do phagocytic cells, such as neutrophils and macrophages, kill intralysosomal pathogens, such as Coxiella burnetii?

Phagocytic cells kill intralysosomal pathogens through oxygen-dependent and oxygen-independent mechanisms, including reactive oxygen species and enzymes such as lysozyme and phospholipases.

What is the role of IcsA in Shigella's actin-based motility?

IcsA recruits N-WASP, which recruits Arp2/3, driving actin polymerisation and promoting bacterial spread.

What is the primary function of Listeria's LLO protein in its actin-based motility?

LLO is a pore-forming toxin that helps Listeria escape the vacuole and enter the cytosol, where it can polymerise actin and promote cell-cell spread.

What is the role of ActA in Listeria's actin-based motility?

ActA mimics WASP, recruiting Arp2/3 and driving actin polymerisation, which promotes bacterial spread.

How do cytosolic pathogens, such as Listeria and Shigella, recruit host cell cytoskeletal proteins to promote actin polymerisation?

Listeria uses ActA to mimic WASP, while Shigella uses IcsA to recruit N-WASP, both of which recruit Arp2/3 to drive actin polymerisation.

What is the primary function of phagocytosis in phagocytic cells, such as neutrophils and macrophages?

Phagocytosis allows phagocytic cells to engulf and eliminate foreign particles, including bacteria, from the body.

What is the primary mechanism by which bacteria sequester metal ions, and how do they adapt to overcome host-imposed nutrient limitations?

Bacteria sequester metal ions through the production of siderophores, which are high-affinity iron chelators. To adapt to host-imposed nutrient limitations, bacteria have evolved mechanisms such as the Znu system, which enables them to overcome zinc limitation.

What is the significance of nicotianamine-like metallophores in bacterial metal transport, and how do they facilitate metal ion release?

Nicotianamine-like metallophores are involved in the transport of metal ions across the bacterial membrane, facilitating metal ion release through imperfect coordination chemistry.

What is the role of the Psa permease in bacterial metal transport, and how does it facilitate metal ion release?

The Psa permease is a bacterial transporter that facilitates metal ion release through imperfect coordination chemistry, allowing bacteria to acquire essential metal ions.

What is the significance of nutritional immunity in the battle for nutrient metals at the host-pathogen interface, and how do bacteria adapt to overcome host-imposed limitations?

Nutritional immunity is a host defense mechanism that restricts the availability of essential metal ions to pathogens, and bacteria adapt to overcome this limitation through the production of siderophores and other metal transport systems.

What is the role of the Acinetobacter baumannii Znu system in overcoming host-imposed nutrient zinc limitation, and how does it facilitate metal ion acquisition?

The Acinetobacter baumannii Znu system is a bacterial transporter that facilitates metal ion acquisition by overcoming host-imposed zinc limitation, allowing bacteria to acquire essential metal ions.

What is the significance of salmochelins in bacterial metal transport, and how do they facilitate metal ion release?

Salmochelins are C-glucosylated enterobactins produced by Salmonella enterica, which facilitate metal ion release through imperfect coordination chemistry, allowing bacteria to acquire essential metal ions.

What is the molecular mechanism by which bacteria are susceptible to zinc, and how does it impact bacterial metal ion acquisition?

Bacteria are susceptible to zinc due to the inhibition of metal ion acquisition by zinc, which disrupts the function of bacterial metal transport systems, such as the Znu system.

What is the role of the psa permease complex in bacterial metal transport, and how does it facilitate metal ion release?

The psa permease complex is a bacterial transporter that facilitates metal ion release through imperfect coordination chemistry, allowing bacteria to acquire essential metal ions.

What is the significance of bacterial iron piracy in the evolution of transferrin, and how does it impact host-pathogen interactions?

Bacterial iron piracy is a mechanism by which bacteria acquire iron from the host, and it has driven the evolution of transferrin as a host defense mechanism, leading to a rapid evolution of transferrin to evade bacterial iron piracy.

What is the role of metallophores in the battle for nutrient metals at the host-pathogen interface, and how do they facilitate metal ion acquisition?

Metallophores are molecules produced by bacteria that facilitate metal ion acquisition by chelating metal ions, allowing bacteria to overcome host-imposed limitations on metal availability.

What mechanism do Neisseria gonorrhoeae use to evade the host's immune response, and what is the result of this process?

Antigenic variation by recombination between different pilin genes, resulting in a new pilin protein on the surface that is not recognized by host antibodies and therefore cannot be easily cleared by the host.

What is the purpose of antigenic variation in bacterial pathogens, and how does it contribute to their ability to evade the adaptive immune response?

The purpose of antigenic variation is to modify the surface proteins of the bacterium, making it unrecognizable to the host's immune system, and allowing it to evade clearance by the adaptive immune response.

How do bacteria modify themselves to avoid clearance by the adaptive immune response, and what is the significance of this process in the context of bacterial pathogenesis?

Bacteria modify themselves using phase and antigenic variation, allowing them to change their surface proteins and evade recognition by the host's immune system. This process is significant in the context of bacterial pathogenesis, as it allows bacteria to evade the host's immune response and cause disease.

What is the role of recombination between pilin genes in the antigenic variation of Neisseria gonorrhoeae?

Recombination between pilin genes results in the creation of new pilin proteins on the surface of N. gonorrhoeae, which are not recognized by the host's immune system.

How do bacteria use antigenic variation to evade the host's immune response, and what are the consequences of this process for the host?

Bacteria use antigenic variation to modify their surface proteins, making it difficult for the host's immune system to recognize and eliminate them. The consequences of this process for the host are the inability to clear the infection, leading to disease.

What is the significance of the evasion of innate immunity by bacteria, and how do they achieve this evasion?

The evasion of innate immunity is significant because it allows bacteria to establish an infection and cause disease. Bacteria achieve this evasion through various mechanisms, including the modification of their surface proteins to avoid recognition by host immune cells.

What is the role of phase variation in the evasion of the adaptive immune response by bacteria?

Phase variation is a mechanism used by bacteria to modify their surface proteins, allowing them to evade recognition by the host's immune system and avoid clearance by the adaptive immune response.

How do bacteria use antigenic variation to evade the host's immune response, and what are the implications of this process for vaccine development?

Bacteria use antigenic variation to modify their surface proteins, making it difficult for the host's immune system to recognize and eliminate them. The implications of this process for vaccine development are that vaccines must be able to target multiple antigenic variants to be effective.

What is the significance of the ability of bacteria to evade the adaptive immune response, and how does this ability contribute to their ability to cause disease?

The ability of bacteria to evade the adaptive immune response is significant because it allows them to establish an infection and cause disease. This ability is critical in the context of bacterial pathogenesis, as it allows bacteria to evade the host's immune system and cause disease.

How do bacteria use antigenic variation to evade the host's immune response, and what are the consequences of this process for the host's immune system?

Bacteria use antigenic variation to modify their surface proteins, making it difficult for the host's immune system to recognize and eliminate them. The consequences of this process for the host's immune system are the inability to clear the infection, leading to disease and potential immune system dysfunction.

What are the limitations of Koch's Postulates in the modern era of molecular biology?

Koch's Postulates do not account for asymptomatic carriers, chronic infections, and non-cultivable microorganisms. They also do not consider the role of environmental factors and the host's immune response.

What is the purpose of the three principles outlined in Koch's Postulates in the molecular era?

To associate a microbial gene with a virulence trait, to demonstrate the loss of virulence when the gene is mutated, and to restore virulence when the gene is restored.

What is the significance of using a model organism in identifying virulence genes?

A model organism allows for the study of a specific virulence trait in a controlled and simplified system, making it easier to identify and characterize the genes involved.

What are the benefits and limitations of the array-based screen used to identify virulence genes?

Benefits: high-throughput, allows for the screening of multiple genes simultaneously. Limitations: may not detect all virulence genes, may not account for gene interactions and regulation.

What are the advantages of using molecular methods over traditional Koch's Postulates in identifying virulence factors?

Molecular methods allow for the identification of virulence genes in non-cultivable organisms, and enable the study of gene regulation and interaction. They are also faster and more efficient than traditional methods.

What is the primary mechanism by which affinity maturation of antibodies occurs in center B cells?

Positive selection, where B cells with higher affinity receptors are preferentially selected and activated

What is the purpose of selective pressure in the evolution of new traits in a population?

To drive the selection of individuals with favorable traits, leading to the adaptation of the population to its environment

What is the primary function of the Sec Translocase pathway in protein secretion?

To translocate unfolded proteins across the plasma membrane, allowing them to be folded and modified in the periplasm

What is the role of the T6SS system in interspecies interactions?

To deliver effector proteins into neighboring bacteria, allowing for the manipulation of their behavior and physiology

What is the primary function of the Type 7 Secretion System (T7SS) in Gram-positive bacteria?

To secrete proteins involved in virulence and pathogenesis, allowing for the establishment of infection

What is the main difference in the mechanism of action between Tetracycline and Linezolid?

Tetracycline binds to the 30S ribosomal subunit, preventing amino-acyl tRNA binding to the A site, whereas Linezolid binds to the 50S ribosomal subunit, blocking the P-site and stopping initiation of protein biosynthesis.

What is the significance of the origin of Tetracycline and Linezolid?

Tetracycline is derived from the bacterium Streptomyces aureofaciens, while Linezolid is a synthetic antibiotic.

How do Tetracycline and Linezolid differ in terms of resistance?

Tetracycline resistance is often due to ribosomal methylation, while Linezolid resistance is generally low due to its unique mechanism of action.

What is the significance of the 30S and 50S ribosomal subunits in the context of Tetracycline and Linezolid?

The 30S subunit is the target of Tetracycline, while the 50S subunit is the target of Linezolid.

What is the primary consequence of the mechanisms of action of Tetracycline and Linezolid?

Both antibiotics ultimately inhibit protein production, leading to bacteriostatic effects.

What is the primary goal of antibiotic stewardship, and how can it be achieved?

The primary goal of antibiotic stewardship is to reduce antibiotic resistance. This can be achieved by using fewer antibiotics, using them only when necessary, and stopping unnecessary use in agriculture.

What are the advantages and disadvantages of phage therapy as an alternative treatment to antibiotics?

The advantages of phage therapy are that it is highly specific and can kill bacteria rapidly. The disadvantages are that it requires lytic phages, is highly specific and therefore requires a cocktail of phages, and is difficult to commercialize.

What is the main problem with finding new antibiotics, and how can it be overcome?

The main problem is that there are no new antibiotic classes since the 1990s. This can be overcome by using alternative approaches such as using natural products, iChips, and induction to access new bacteria and silent genes.

What is the significance of natural products in antibiotic discovery, and what are the challenges associated with their use?

Natural products, primarily from Streptomyces, represent over 70% of antibiotics in clinical use. The challenges are that identifying new structures is difficult and complex structures are not easily modified or synthesized.

What are the two key problems with current antibiotic discovery, and how can they be addressed?

The two key problems are rediscovery of same compounds and difficulty in accessing chemical diversity. These can be addressed by using new methods such as iChips, induction, and cloning and expressing genes.

What is the significance of the golden age of antibiotics, and what has happened since then?

The golden age of antibiotics was a period of significant antibiotic discovery. However, since the 1990s, there have been no new antibiotic classes, highlighting the need for new approaches to antibiotic discovery.

What is the goal of whole cell assays in antibiotic discovery, and how does it differ from target-based screening?

The goal of whole cell assays is to kill bacteria, while target-based screening aims to inactivate enzymes. Whole cell assays require knowing the molecular target and cell entry, whereas target-based screening requires knowing the molecular target.

What is the significance of the case study of GSK's library screening, and what does it reveal about the challenges of antibiotic discovery?

The case study reveals that despite a significant investment of time and resources, only a few leads were identified, and none resulted in actual products. This highlights the challenges of antibiotic discovery.

What are the mechanisms of antibiotic resistance, and why is it a complex problem?

The mechanisms of antibiotic resistance include efflux, inactivation, and target modification. Antibiotic resistance is a complex problem because it involves multiple factors and is a multifactorial issue.

What is the significance of antibiotic resistance, and why is it a pressing issue?

Antibiotic resistance is a significant public health issue because it makes infections harder to treat, leading to increased morbidity and mortality. It is a pressing issue because it requires urgent action to combat it.

What is the primary feature of clade A E. faecium that distinguishes it from clade B?

Altered cell wall and capsule

What is the mechanism by which E. faecium primarily emerges to cause hospital-acquired infections?

Gaining genes through horizontal gene transfer (HGT)

What is the characteristic of E. faecium genome that allows for the dynamic acquisition of new genetic material?

Constant flux of accessory genes through HGT events

What is the primary method of acquiring new genetic material in E. faecium?

Conjugation

How many unique antibiotic resistance genes are found in E. faecium genomes?

34

What is the average number of antibiotic resistance genes found in E. faecium genomes?

9

What is the reason for E. faecium's intrinsic resistance to most β-lactam antibiotics?

Low affinity for PBP5

What is the cure rate of monotherapy for endocarditis treatment using ampicillin?

< 40%

What is the mechanism by which E. faecium gains antibiotic resistance genes?

Horizontal gene transfer

What is the significance of E. faecium's ability to adapt to the hospital environment?

Better adaptation to the hospital environment

What percentage of CF patients have lung infections with P. aeruginosa by adulthood?

Up to 80%

What is a characteristic of P. aeruginosa strains isolated from CF lungs that is rarely seen in individuals without CF?

Dramatic genetic, morphological, and physiological changes

What is a mechanism of antibiotic resistance in P. aeruginosa and A. baumannii?

Multidrug efflux pumps

What is the primary function of the Resistance-Nodulation-Division (RND) family of efflux pumps?

Proton/substrate antiporters

How many RND transport systems are found in P. aeruginosa?

13

What is the name of the RND systems involved in drug efflux in A. baumannii?

AdeABC, AdeFGH, and AdeIJK

What is the primary consequence of the low antibiotic susceptibility of P. aeruginosa and A. baumannii?

Limited treatment options

What is the primary factor contributing to the decrease in the pipeline of new antibiotics?

Multifactorial resistance

What is the primary mechanism of antibiotic inactivation or modification in P. aeruginosa and A. baumannii?

Antibiotic-degrading and antibiotic-inactivating enzymes

What is the primary function of the biofilm barriers in P. aeruginosa and A. baumannii?

Protection from antibiotics

What is the difference between colonization and infection, and why is it important in the context of hospital-acquired infections?

Colonization refers to the presence of microorganisms on or inside the body without causing harm, whereas infection refers to the multiplication of microorganisms that cause harm to the host. Understanding the difference is crucial in hospital-acquired infections to develop effective prevention and treatment strategies.

What are some characteristics of high-scoring answers in short-answer questions, and why are they important?

High-scoring answers typically provide clear explanations, demonstrate understanding of the question and response, and support opinions with facts. They are important because they showcase a student's ability to think critically and communicate effectively.

What is the significance of genome sequencing in tracking disease transmission, and how does it aid in hospital-acquired infection control?

Genome sequencing enables the tracking of disease transmission by identifying the genetic makeup of microorganisms, allowing for targeted interventions. It aids in hospital-acquired infection control by identifying the source of infections and implementing effective prevention strategies.

What are some common pitfalls to avoid when answering short-answer questions, and why are they important to consider?

Common pitfalls to avoid include brain dumping, failing to read the question carefully, and providing limited or unsupported answers. These mistakes can result in lost marks and misunderstandings.

What are some key features of hospital-acquired infection control, and why are they essential for patient safety?

Key features of hospital-acquired infection control include surveillance, isolation precautions, and sterilization. They are essential for preventing the spread of infections and ensuring patient safety.

What limitations do the authors of the study on hospital-acquired infections highlight, and how do these limitations impact the reliability of their findings?

The authors highlight limitations such as differences in surveillance methods, regional differences in data sources, and limited evidence of clonal relationship between colonizing and infecting strains. These limitations impact the reliability of their findings by introducing potential biases and reducing the accuracy of their conclusions.

What are the key factors that need to be considered when tracking the spread of a Gram-positive organism within a hospital, and how would you determine its origin?

Key factors to consider include the origin of the isolates, patient demographics, and potential transmission routes. To determine the origin, one would need to compare the genetic sequences of the isolates to identify any clonal relationships and conduct phylogenetic analysis.

What are the potential risk factors that could contribute to the spread of a Gram-positive organism within a hospital, and how would you design a study to investigate these factors?

Potential risk factors include inadequate hand hygiene, contaminated surfaces, and improper use of personal protective equipment. A study to investigate these factors could involve a combination of epidemiological and molecular analysis, including whole-genome sequencing and phylogenetic analysis.

What are the implications of MDR S. epidermidis being a potential problem in hospitals, and what are the potential mechanisms of resistance to vancomycin in this organism?

The implications of MDR S. epidermidis being a potential problem in hospitals include the increased risk of treatment failure and poor patient outcomes. The potential mechanisms of resistance to vancomycin in this organism include the production of vancomycin-modifying enzymes and alterations in the bacterial cell wall.

How would you design a study to investigate the potential for clonal expansion of MDR S. epidermidis in a hospital, and what are the implications of such expansion for patient care and infection control?

A study to investigate the potential for clonal expansion of MDR S. epidermidis could involve a combination of epidemiological and molecular analysis, including whole-genome sequencing and phylogenetic analysis. The implications of clonal expansion include the increased risk of transmission and colonization of patients, highlighting the need for targeted interventions to prevent and control hospital-acquired infections.

What is the effect of diphtheria toxin and Pseudomonas exotoxin A on protein synthesis?

Inhibits protein synthesis

What is the function of Edema Factor (EF) in Anthrax toxin?

Adenylate cyclase

What is the effect of Lethal Factor (LF) in Anthrax toxin?

Inactivates MAPKK leading to apoptosis and death

What is the function of Protective Antigen (PA) in Anthrax toxin?

B component

What is the significance of Anthrax toxin being a multi-chain A-B exotoxin?

It has three proteins: PA, EF, and LF

What is the difference between Anthrax toxin and other exotoxins?

Not all Exotoxins are Proteins

What is the effect of Cholera toxin, TEC heat-labile toxin, and Pertussis toxin on adenylate cyclase?

Activates adenylate cyclase

What is the effect of C.botulinum C2 toxin and CDT binary toxin on actin polymerisation?

Inhibits actin polymerisation

What is the effect of S.aureus EDIN toxin on the Golgi?

Disassembles Golgi

What is the significance of the 'A' and 'B' components of Anthrax toxin?

PA is the 'B' component, EF & LF are 'A' components

What is the main cause of tight junction damage and disruption of epithelial permeability barrier in botulism?

Botulinum toxin (BoNT)

What is the primary target of botulinum toxin (BoNT) in the human body?

The nervous system

What is the most common form of botulism in the US?

Infant botulism (floppy baby syndrome)

What is the mechanism by which botulinum toxin enters neurons?

Endocytosis at presynaptic membrane

What is the result of botulinum toxin's action on nerve function?

Blocks release of acetylcholine into synaptic cleft

What is the purpose of the light chain in botulinum toxin?

Cleaves SNARE-family proteins by proteolysis

What is the purpose of the heavy chain in botulinum toxin?

Binding to neurons

What is the significance of the 7 distinct BoNT types (A-G) in human disease?

Types A,B,E and rarely F are associated with human disease

What is the mode of transmission of botulism in wound botulism?

Contamination of wound with C. botulinum

What is the potency of botulinum toxin compared to other biological toxins?

The most potent biological toxin known

What is the main basis for differentiating between subspecies of T. pallidum, such as pertenue and yaws?

Clinical features of disease

What is the size of the genomes of all subspecies of T. pallidum?

Approximately 1.14 Mbp

What is the percentage of difference at the nucleotide level between all three subspecies of T. pallidum?

0.2%

What is the primary adaptation of T. pallidum to mammalian tissues?

Limited metabolic capacity and many genes for transporters

What is the main method of detection for syphilis?

Serology

What is the purpose of darkfield microscopy in the diagnosis of syphilis?

To detect T. pallidum directly

What is the primary limitation of point-of-care tests for syphilis?

Cannot differentiate between prior treated and current infection

What is the historical treatment for syphilis?

Salvarsan

Why is T. pallidum a human-restricted pathogen?

Humans are the only reservoir of T. pallidum

What is the significance of the lack of oxidative phosphorylation in T. pallidum?

No need for iron or cytochromes

What are the common symptoms of gastrointestinal (GI) infections?

Diarrhea (≥ 3 loose stools over a 24hr period), abdominal pain, vomiting, fever, nausea, and dehydration.

What is the significance of whole-genome sequencing in diagnosing Shigella infections?

Conventional bacterial culture is still the gold standard for diagnosing Shigella infection, as modern techniques like MALDI-TOF cannot distinguish Shigella from pathogenic E. coli.

What is the relationship between Shigella and E. coli?

Shigella is regarded as a pathogenic biotype of E. coli, and the four species of Shigella would be considered serologically defined anaerogenic biotypes of E. coli.

What is the significance of diarrhea diseases in terms of mortality?

Diarrheal diseases are a leading cause of morbidity and mortality, with 1.31 million deaths, and 38.1% of these deaths occur in children.

What are some common causes of gastrointestinal (GI) infections?

Rotavirus, Shigella, and Salmonella are some of the most common causes of GI infections.

What is the likely origin of Shigella clusters, except for S. boydii 13?

Non-invasive E. coli

What is the typical infectious dose of Shigella, and what is the primary symptom of Shigellosis?

10-100 bacteria, diarrhea

What are the common modes of transmission for Shigella?

Person-to-person, contaminated food or water, sexually transmitted

Approximately how many cases of Shigellosis were estimated to occur in 2017?

188 million

Why is the original taxonomy of Shigella still used today?

It remains in line with clinical diseases

What are the common symptoms of gastrointesinal (GI) infections?

Diarrhea (≥ 3 loose stools over a 24hr period), abdominal pain, vomiting, fever, nausea, and dehydration

What is the significance of Shigella and Salmonella in diarrheal diseases?

They are the most common aetiologies, with Shigella flexineri being more common than Shigella sonnei

Why is conventional bacterial culture still the gold standard for diagnosing Shigella infection?

It is not easy to distinguish Shigella from other enteric bacteria using methods like MALDI-TOF

What is the relationship between Shigella and E. coli?

Shigella can be regarded as a serologically defined anaerogenic biotype of E. coli

What is the role of the type 3 secretion system (T3SS) in Salmonella and Shigella?

It plays a key role in the invasive mechanisms of these bacteria, facilitating gut invasion and intracellular survival

Based on genotypic diversification, what is the likely origin of Shigella clusters?

Non-invasive E. coli by convergent evolution

What is the primary symptom of Shigellosis, and what is the range of clinical presentations?

Primary symptom is diarrhoea, ranging from asymptomatic, intestinal to invasive

How is Shigella typically transmitted, and what are the estimated cases of Shigellosis?

Transmitted through person-to-person contact, contaminated food or water, and sexually; estimated 188 million cases in 2017

What is the significance of Shigella being classified as EIEC (Enteroinvasive E. coli)?

It indicates that Shigella emerged from non-invasive E. coli by convergent evolution

Why is Shigella still classified under its original taxonomy, despite its genetic similarity to E. coli?

To maintain consistency with clinical diseases

How can genomics be used to improve vaccine design for Group A Streptococcus?

By assessing 'naturally' occurring genetic variation through population genomics, which allows for the identification of genetic differences between strains and the functional attributes of major virulence factors.

What are some key virulence factors of Group A Streptococcus?

Major contributors to genetic differences between strains, including functional attributes of major virulence factors.

How can genomic epidemiology be used to understand the emergence of pathogenic clones of S.pyogenes?

By identifying molecular markers and characterizing bacterial outbreaks through genomic analysis.

What are the steps in using genomics to characterize bacterial outbreaks?

Sequence, assemble, blast, and output, as illustrated in the Davies et al. study.

Why is Group A Streptococcus a major human pathogen?

Due to its key epidemiological differences and virulence factors.

What are some applications of genomics in understanding bacterial evolution?

Identifying genetic variation, tracking disease transmission, and informing public health interventions.

How can genomics be used to develop new antibiotics?

By identifying novel targets for antibiotic development and informing the design of new antibiotics.

What is the significance of monitoring how pathogen populations respond to interventions?

It allows for the assessment of the effectiveness of interventions and the identification of emerging threats.

How can genomics be used to design public health interventions?

By identifying genetic differences between strains, informing vaccine design, and tracking disease transmission.

What is the focus of genomic epidemiology?

The study of genetic variation at a population level to inform public health interventions.

What are the two main diagnostic tools for active tuberculosis disease?

Microscopy and culture

What is GeneXpert, and what is its significance in tuberculosis diagnosis?

A cartridge-based PCR test

What is the typical treatment regimen for tuberculosis, and what is the duration?

2 months of isoniazid/rifampicin/pyrazinamide/ethambutol followed by 4 months of isoniazid/rifampicin

What is multidrug-resistant tuberculosis (MDR-TB), and how many cases were reported in 2018?

Resistant to isoniazid and rifampicin

What is extensively drug-resistant tuberculosis (XDR-TB), and how many cases were reported in 2018?

Resistant to isoniazid and rifampicin, a fluoroquinolone, and an aminoglycoside

What is the approximate number of new tuberculosis cases reported annually?

10 million

What is the significance of line-probe assays (LPAs) in tuberculosis diagnosis?

Detect multiple resistance markers

Why is there a need for new and repurposed 'old' drugs in tuberculosis treatment?

Emergence of drug-resistant variants

What is the current limitation in tuberculosis diagnosis, especially in resource-limited settings?

Lack of simple, effective, and rapid point-of-care (POC) tests

What is the significance of tuberculosis treatment in terms of duration and multiple drugs?

Requires simultaneous administration of multiple drugs over a long period

What is the common factor among hospitals where cases of mycobacterial infections occurred across geography and time?

All hospitals had used one brand of HCU

What is the mechanism of action of isoniazid in mycobacterial cells?

Binds tightly to InhA, blocking fatty acid synthesis and mycolic acid synthesis

What is the primary target of ethambutol in mycobacterial cells?

Arabinosyl transferase (embCAB operon)

What is the unique mechanism of action of bedaquiline?

Inhibits mycobacterial ATP synthase

What is the primary function of pyrazinamide in mycobacterial infections?

Active against persister cells

What is the role of delaminid/pretomanid in mycobacterial infections?

Inhibits cell wall synthesis, blocking mycolic acid synthesis

What is the target of telacebec (Q203) in mycobacterial infections?

Cytochrome bc1

What is the duration of treatment for mycobacterial infections using rifampicin/ethambutol/a macrolide?

12 months+

What is the significance of the chimaera containing aerosols in mycobacterial infections?

Contaminated operating field

What is the focus of anti-mycobacterial therapy in mycobacterial infections?

Bactericidal in rapid growers, bacteriostatic in slow growers

What was the mortality rate in untreated patients with pneumococcal bacteraemia?

80%

What was the mortality rate among patients who received treatment with antibiotics such as penicillin or amoxycillin?

17%

What percentage of isolates have been reported to show multi-drug resistance?

up to 46%

What is the treatment failure rate due to antibiotic resistance?

up to 30%

What is one approach to addressing antibiotic resistance in Streptococcus pneumoniae?

Developing new antimicrobials targeting highly conserved pathways and proteins

Why is Streptococcus pneumoniae considered a very good human pathogen?

Due to its ability to cause severe disease and high mortality rates

What is the significance of evolutionary pressures exerted by the human host on Streptococcus pneumoniae?

It may be the Achilles' heel of the pneumococcus

What is the current situation regarding antibiotic treatment of Streptococcus pneumoniae infections?

Treatment failure occurs in up to 30% of cases due to antibiotic resistance

What is the relationship between Streptococcus pneumoniae and other major diseases?

S. pneumoniae kills more people per year than AIDS, TB, and malaria combined

What is the significance of novel therapeutics in combating Streptococcus pneumoniae infections?

They are needed to address the rising antibiotic resistance

How do Burkholderia species modify their Lipid A to resist the activities of cationic peptides?

By chemically altering their Lipid A

What is the significance of protein glycosylation in Burkholderia species?

It is required for virulence

What is the impact of O-linked glycosylation on virulence across Burkholderia species?

It is a critical factor

How do sugars contribute to the virulence of Burkholderia species?

They can be used to mask or block access to features at the bacterial surface

What is the role of the humoral response in detecting exposure to Burkholderia species?

It is used to detect exposure

What is the significance of protein glycosylation in Burkholderia species in relation to immunogenicity?

It is highly immunogenic

How do Burkholderia species use glycosylation to impact virulence attributes?

Glycosylation impacts virulence attributes

What is the role of carbohydrates in shielding or blocking access to features at the bacterial surface?

They can be used as structural building blocks

What is the relationship between protein glycosylation and virulence in Burkholderia species?

Glycosylation is required for virulence

What is the significance of glycans in Burkholderia species in relation to immune detection?

Glycans can be used to detect exposure

In the context of the 1976 Legionnaires' disease outbreak, what would be the primary method of identifying the causative agent in the absence of genomics and PCR?

Gram stain, animal models, and antisera

What type of bacterium was identified as the cause of Legionnaires' disease in 1977?

Gram negative bacterium

What would you need to pack in the Tardis to investigate the outbreak in 1976?

Equipment and reagents for Gram stain, animal models, and antisera

What was the mortality rate of the initial infected cohort in the 1976 Legionnaires' disease outbreak?

16%

What was the significance of the 1976 Legionnaires' disease outbreak?

Identification of a new disease and its causative agent

What was the time frame between the outbreak and the publication of the papers outlining the disease?

One year and five months

What was the location of the State Convention of Legionnaires where the outbreak occurred?

The Bellevue-Stratford Hotel in Philadelphia, Pennsylvania, USA

What was the method of transmission of the disease according to the initial outbreak?

Respiratory tract

What was the total number of cases reported in the initial outbreak?

182

What was the significance of the Tardis in the context of investigating the 1976 outbreak?

A machine that enables time travel, allowing the investigator to go back to 1976 with modern equipment and reagents

Study Notes

Genomics, Microbial Evolution, and Epidemiology

  • Definition of a microbial genome: the total genetic content of an organism
    • In the context of bacteria, it includes the chromosome(s) and plasmid(s)
  • Definition of genomics: the study of the structure and function of genes, including the analysis of genome sequences and their applications

Sequencing a Microbial Genome

  • Four-step process:
    1. Generate sequencing reads
    2. Read-mapping or de novo assembly
    3. Finishing (most accurate method is to PCR sequence across gap regions or use a different platform)
    4. Genome annotation
  • DNA sequencing technologies:
    • Oxford Nanopore: single molecule, real-time sequencing
    • Illumina: genome analyser
  • Genome sequencing "explosion" due to the rapid decrease in sequencing cost and increase in speed

Genome Annotation

  • Process of locating the position of genes in the genome
  • Identifies each open reading frame (ORF) in the genome
  • ORF: a reading frame >100 codons that is not interrupted by a stop codon
  • CDS: protein-coding DNA sequence encoded by a gene
  • BLAST (basic local alignment search tool) computer program:
    • Base-by-base comparison of two or more gene sequences
    • Assigns tentative function of gene or protein structure based on BLAST alignment
  • Gene function is often inferred from similarity to other genes, but experimental evidence is rarely used to support annotation

Bioinformatics

  • Analysis of genomic data using computers
  • Addresses novel questions in microbiology, including:
    • Disease transmission and spread
    • Evolution, including drug resistance
    • Pathogenesis and virulence
    • Microbial ecology

Applications of Genomics

  • Foundation for other omic studies (transcriptome, proteome, etc.)
  • Rational vaccine design: identifying potential vaccine targets
  • Novel drug targets: identifying genes/pathways essential for bacterial growth
  • DNA sequence-based diagnostics
  • Understand variation in populations: population genetics, phylogenetics, and phylogeography

Bacterial Evolution and Population Genetics

  • Identifying mutations within a population
  • Assessing genetic variation at a population level
  • Determining alleles at single nucleotide loci (SNPs)
  • Mapping sequence reads to a reference genome

Phylogenetics and Phylogeography

  • Inferring phylogeny from a set of genomic sequences
  • Estimating evolutionary relationships among a set of genome sequences
  • Phylogeography: combining phylogenetics with geographical mapping to infer how bacteria spread
  • Applications in epidemiology: following disease transmission and spread

Epidemiology Revision

  • Definition: the science that evaluates the occurrence, determinants, distribution, and control of health and disease in a defined human population
  • "Genomic epidemiology" links genomics with epidemiology
  • Example: contact tracing

Transport Systems in Gram-Negative and Gram-Positive Bacteria

  • Two common transport systems found in both Gram-Negative and Gram-Positive bacteria are the General Secretory Pathway (Sec Translocase Pathway) and the Twin Arginine Transport (TAT) pathway.
  • The Sec Translocase Pathway is responsible for the secretion of unfolded proteins, and can be a co-translational or post-translational process.
  • Proteins targeted for the Sec system have a signal tag, a 20-30 residue sequence, at the N-termini.
  • The TAT system allows the export of folded proteins from the cytoplasm, recognizing a twin arginine tag at the N-termini of proteins.

Transport Across the Inner Membrane

  • The Sec Translocase Pathway allows the transport of unfolded proteins across the inner membrane.
  • The TAT system allows the export of folded proteins from the cytoplasm, recognizing a twin arginine tag at the N-termini of proteins.

Secretion into the Environment

  • The Type 5 Secretion System (T5SS) allows the translocation of proteins across the plasma membrane after Sec translocation.
  • T5SS proteins contain four key elements: an N-terminal signal tag, an internal passenger domain, a linker region, and a C-terminal β-barrel/translocator domain.
  • Insertion of the translocator domain is mediated by the BAM complex within the outer membrane.
  • Multiple classes of T5SS exist, some anchor proteins to the surface, and some allow the release of proteins after cleavage.

T1SS and T5SS Proteins Associated with Virulence

  • T1SS: α-haemolysin (HlyA of E. coli) is associated with virulence.
  • T5SS: IgA1 protease of Neisseria Gonorrhoeae is associated with virulence.
  • T5SS: Ag43 of E. coli is associated with virulence.

Transport Systems Which Translocate Proteins into Hosts

  • Some Gram-Negative pathogens have evolved dedicated secretion systems that can translocate proteins across 3 membranes (inner/outer/host).
  • These secreted proteins are commonly referred to as 'Effectors'.
  • The Type 4 Secretion System (T4SS) enables the translocation of DNA or proteins to other bacteria as well as eukaryotic cells.
  • T4SS has two main subfamilies: conjugation machineries (DNA and protein transfer via the VirB/VirD system-T4ASS) and effector protein translocation systems (protein transfer via Dot/Icm system T4BSS).

T4SS Subfamilies

  • The VirB/VirD system is evolutionarily related to bacterial conjugation systems.
  • The Dot/Icm system is responsible for the translocation of 100s of proteins that block, blunt, and subvert host cells.

Secretion into Other Cells: T6SS

  • T6SS appears to share structural features with bacteriophage cell puncturing proteins.
  • T6SS is a contact-dependent secretion mechanism that acts as a "poisoned spear" to deliver effectors.
  • T6SS targets both bacteria and eukaryotic cells.
  • Found in ~25% of Gram-negative bacteria, including human and animal pathogens.

T6SS, Inter-Bacterial Combat

  • T6SS can be used as a mechanism to compete with other bacteria (bacterial antagonism).
  • Injection (stabbing) of toxins into neighbouring bacterial cells.
  • 'Attacker' is protected via cognate immunity proteins.
  • Can also be used as a form of bacterial retaliation (tit for tat phenomena).

Transport Systems Unique to Gram-Positive Bacteria (T7SS)

  • Unique secretion systems exist in Gram-Positive bacteria, such as the Type 7 Secretion system (T7SS).
  • Prototypic example from Mycobacterium tuberculosis (related systems also seen in Staphylococcus aureus).
  • T7SS transports proteins across the inner membrane and the mycobacterial cell wall, dependent on a C-terminal signal tag.
  • Required for virulence.

Summary

  • Multiple secretion systems exist (T1SS, T3SS, T4SS, T5SS, T6SS, and T7SS).
  • Systems differ in their ability to transport folded/unfolded proteins, signal sequence locations (N and C termini), and target location (host and environment).

Sec and Tat Secreted Proteins and Virulence

  • Sec and Tat secreted proteins can contribute to virulence in bacteria.
  • Example: NamA autolysins of Listeria monocytogenes contribute to host colonization by remodeling peptidoglycan.
  • Example: Phospholipase C of Pseudomonas aeruginosa (PlcH) requires Tat-based secretion to be functional.

Transport Systems in Gram-Negative Bacteria

  • Gram-negative bacteria have two membranes (inner and outer) that require unique mechanisms for protein secretion.
  • Secretion systems in Gram-negative bacteria can be divided based on where they deposit their proteins.
  • There are multiple transport systems that allow the transport of proteins across the outer membrane from both the cytoplasm and periplasmic space.

Types of Secretion Systems

  • Type I secretion (T1SS): translocates proteins across the outer membrane in a one-step process.
  • Type II secretion (T2SS): translocates proteins across the outer membrane in a multi-step process.
  • Type III secretion (T3SS): translocates effectors directly into the cytoplasm of target eukaryotic cells.
  • Type IV secretion (T4SS): enables the translocation of DNA or proteins to other bacteria or eukaryotic cells.
  • Autotransporter pathway (T5SS): translocates proteins across the outer membrane using a multi-step process.

Transport Systems that Translocate Proteins into Host Cells

  • Type III secretion (T3SS) translocates effectors directly into the cytoplasm of target eukaryotic cells.
  • Type IV secretion (T4SS) can translocate effectors into host cells.
  • Type VI secretion (T6SS) can also translocate proteins into host cells.

Characteristics of Each Secretion System

  • T1SS: C-terminal secretion signal, no folded substrate, 2 membranes, found in E. coli and Pseudomonas.
  • T2SS: N-terminal secretion signal, folded substrate, 1 membrane, found in Aceintobacter.
  • T3SS: N-terminal secretion signal, no folded substrate, 2-3 membranes, found in EPEC, Shigella, Salmonella.
  • T4SS: C-terminal secretion signal, no folded substrate, 2-3 membranes, found in Legionella, Coxiella.
  • T5SS: N-terminal secretion signal, no folded substrate, 1 membrane, found in Neisseria, E. coli.
  • T6SS: secretion signal unknown, partially folded substrate, 2-3 membranes, found in Burkholderia.

Examples of T1SS and T5SS Proteins Associated with Virulence

  • T1SS: α-haemolysin (HlyA of E. coli).
  • T5SS: IgA1 protease (Neisseria gonorrhoeae), Ag43 of E. coli.

Type III Secretion System (T3SS)

  • Found in Gram-negative pathogens, including Salmonella and Yersinia.
  • Translocates effectors directly into the cytoplasm of target eukaryotic cells.
  • Shares homologues with flagella biosynthesis and assembly systems.
  • Effector proteins contain an N-terminal sequence recognized and bound by specific chaperones.
  • Chaperones guide effector proteins to the secretion apparatus.

Type IV Secretion System (T4SS)

  • Enables the translocation of DNA or proteins to other bacteria or eukaryotic cells.
  • Two main subfamilies: conjugation machineries (DNA and protein transfer) and effector protein translocation systems.
  • Examples: Helicobacter pylori Cag system and Bordetella pertussis pertussis toxin secretion.

Bacterial Outer Membrane Vesicles (OMVs)

  • OMVs can enter human epithelial cells via multiple mechanisms to interact with NOD1 and be cleared by the host
  • OMVs can activate other host pathogen recognition receptors (PRRs) to mediate inflammation and pathogenesis in the host

Pathogen Recognition Molecules (PRMs)

  • The TLR family of bacterial pathogen recognition molecules (PRMs) includes NOD1/2 and TLR9
  • Bacterial products recognized by PRMs include peptidoglycan (PG) and lipopolysaccharides (LPS)

Biogenesis of OMVs

  • OMVs can be produced through mechanisms such as blebbing, bacterial lysis, and membrane remodeling
  • The Tol-Pal system maintains bacterial cell membrane integrity during OMV biogenesis
  • The Tol-Pal cluster consists of inner membrane proteins (TolA, TolQ, TolR), periplasmic protein (TolB), and an outer membrane peptidoglycan-associated protein (Pal)

Functions of OMVs

  • OMVs can mediate inflammation and pathogenesis in the host by activating host innate immune receptors (PRRs)
  • OMVs can deliver virulence determinants to and into host cells to promote pathogenesis
  • Non-pathogenic functions of OMVs include promoting bacterial survival, acquiring nutrients, degrading antibiotics, and transferring resistance

Gram Positive and Gram Negative Bacteria

  • Both Gram positive and Gram negative bacteria produce OMVs with varied cargo
  • OMVs produced by Gram positive bacteria contain cargo that can mediate inflammation and pathogenesis in the host
  • Examples of Gram positive bacteria that produce OMVs include Bacillus anthracis and Mycobacterium tuberculosis

Environmental Factors and OMV Production

  • Environmental factors can increase OMV production by bacteria

Lecture Outcomes

  • Bacterial OMVs are a novel secretion system used by all bacteria
  • OMVs have different structures and cargo in Gram positive and Gram negative bacteria
  • Environmental factors can increase OMV production
  • OMVs can mediate inflammation in the host by activating host innate immune receptors (PRRs)

Diagnostic Microbiology

  • Diagnostic microbiology involves using a range of tests to identify the disease-causing agent.
  • The type and handling of clinical specimens are important considerations, as they can affect the accuracy of test results.

Culture Dependent Methods

  • Bacterial culture involves the growth of bacteria on physical media, such as liquid or agar plates.
  • Growth can depend on factors such as temperature, oxygen levels, time, and media type.
  • Selective/differential media are used to isolate or differentiate bacteria from other organisms within a sample.
  • Examples of selective/differential media include EMB agar, which inhibits Gram-positive bacteria and differentiates between lactose fermenters, and Baird-Parker agar, which identifies Staphylococcus aureus.

Biochemical Tests

  • Biochemical tests are based on the differential features of closely related organisms or those from similar sites.
  • Most biochemical tests are based on enzyme production or activity, such as catalase, oxidase, and urease.
  • Other tests involve the utilization of carbohydrates or tolerance to particular conditions, such as 6.5% salt.
  • Biochemical tests are often rolled into kits for specific organisms, such as API test strips.

Automated Culture Systems

  • Automated culture systems, such as BACTEC or BACT/ALERT, are used for blood, sterile fluids, and platelets.
  • These systems grow and scan bottles for bacterial growth, alerting when growth is detected.
  • VITEK 2 is an automated system that provides both identification and antibiotic susceptibility testing.

Culture Independent Detection Methods

  • Microscopy of specimens can be used for direct detection, including staining and direct and indirect immunofluorescence.
  • Immune reactions, such as ELISA, can also be used for detection.
  • Molecular methods, including PCR, DNA sequencing, and MALDI, can be used for direct detection.

Microscopic Detection

  • Direct examination of smears by staining can be used for diagnosis, with a sensitivity of 36.8% for TB diagnosis.
  • Immunofluorescence involves the use of fluorescent antibodies that bind directly to the pathogen, which can be viewed under a microscope.

Molecular Detection Methods

  • PCR and real-time PCR are used for nucleic acid amplification, often where organisms are difficult to culture or speed of diagnosis is essential.
  • Automated systems, such as GeneXpert for TB diagnosis, are available for molecular detection.

Importance of Diagnosis

  • Good diagnosis relies on a combined understanding of the disease and correct interpretation of results.
  • Diagnostic tests can be classified into culture-dependent and culture-independent methods.

CRISPR-Cas Systems

  • CRISPR-Cas systems consist of three essential components: crRNA, trans-activating crRNA, and Cas9 endonuclease.
  • CRISPR-Cas is a class 2, type II system.
  • CRISPR-Cas systems have three stages: adaptation, biogenesis, and interference.

Three Stages of CRISPR Immunity

  • Adaptation (immunisation): the process of acquiring new spacers from invading viruses or plasmids.
  • Biogenesis (crRNA maturation): the process of maturing crRNA from precursor RNA.
  • Interference (immunity): the process of cleaving target DNA using Cas9 endonuclease.

The PAM Sequence

  • PAM (Proto-spacer Adjacent Motif): a sequence present on invading viral or plasmid DNA, but not part of the CRISPR array.
  • PAM sequence is essential for Cas9 to recognise the target sequence and cleave it.
  • The canonical PAM sequence is 5'-NGG-3', but different Cas9 proteins recognise different PAMs (2-6 base pairs).

Bacteria and CRISPR-Cas Systems

  • CRISPR-Cas systems prove that prokaryotic genomes are environmentally tuned.
  • Bacteria can adapt by Lamarckian inheritance, keeping track of and inheriting genomic encounters.
  • CRISPR-Cas systems suggest that there are many more unknown processes yet to be uncovered in the ~30% of function unknown genes in each prokaryotic genome.

Application of CRISPR

  • CRISPR-Cas systems can be harnessed as a genome engineering tool for precise DNA cleavage.
  • Applications of CRISPR include infectious disease, functional genomics, and large-scale genetic screens.
  • CRISPR-Cas systems have been used to develop new cancer therapies and may enable the curing of inherited diseases.

History of CRISPR Discovery

  • CRISPR was first discovered in the 1980s and 1990s as repeat regions in DNA sequences in different microbes.
  • The term "CRISPR" was coined in 2002.
  • The 2020 Nobel Prize in Chemistry was awarded to Emmanuelle Charpentier and Jennifer A. Doudna for the development of CRISPR-Cas9 genome editing.

Anti-CRISPR Mechanisms

  • Anti-CRISPR mechanisms include mutation leading to mismatches and inefficient cleavage, recombination in polylysogenic genomes, anti-CRISPR proteins (e.g. Acr), and phage carrying CRISPR-Cas elements.
  • These mechanisms can inhibit the activity of CRISPR-Cas systems.

Bacterial Encounters

  • Humans encounter millions of bacteria daily, but most interactions are transient
  • The body is a hostile environment for bacteria, with environmental conditions like skin and intestine, mechanical factors like shedding and peristalsis, and a normal flora that makes up 50% of the body's cells
  • The immune system also plays a role in preventing bacterial colonization

Molecular Basis of Bacterial Adhesion

  • Bacterial adhesion is the process by which bacteria attach to host cells or receptors
  • Adhesins are molecules that mediate adherence, including pili/fimbrial and afimbrial adhesins
  • Pili/fimbrial adhesins are complex multigene assemblies with repeating subunits, a tip protein, and a secretion system
  • Examples of pili/fimbrial adhesins include type I pili (fim), pap, and afa/Dr
  • Afimbrial adhesins are single genes with multiple domains, including trimeric autotransporter adhesins (TAAs)

Adhesin Diversity and Immune Evasion

  • Pathogens use alternate colonisation factors, additional domains, and anti-immune mechanisms to evade the immune system
  • Adhesins are not always expressed and are tightly regulated to reduce exposure to the immune system
  • Phase variation and random expression of adhesins also contribute to immune evasion

Escherichia coli Adherence

  • Escherichia coli is a Gram-negative bacterium that causes diarrheal disease
  • Different pathotypes of E. coli have different adhesin complements, including F-pili, AAF, and dispersin

Pathotypes and Adhesins

  • Enteropathogenic E. coli (EPEC) uses Fim, Bfp, LEE, Paa, and LPF adhesins
  • Enterotoxigenic E. coli (ETEC) uses Fim, TCP, and CFAs adhesins
  • Enterohaemorrhagic E. coli (EHEC) uses Fim, LEE, Paa, ToxB, Efa-1/LifA, and LPF adhesins
  • Other pathotypes of E. coli use different combinations of adhesins

Bacterial Invasion and Dissemination Strategies

  • Bacterial invasion involves two main mechanisms: zipper and trigger mechanisms
  • The zipper mechanism exploits host cell pathways normally used for adhesion of epithelial cells to the extracellular matrix
  • The trigger mechanism resembles cell ruffling induced by growth factors and hormones

Zipper Mechanism - Listeria monocytogenes

  • Listeria monocytogenes is a Gram-positive, foodborne pathogen that causes CNS infections and maternofetal infections
  • Internalization requires the actin cytoskeleton and bacterial protein internalin A (InlA)
  • InlA interacts with E-cadherin, which is involved in tight junction formation, leading to receptor clustering and cytoskeletal changes

Zipper Mechanism - Complications

  • Listeria monocytogenes has two invasins: InlA and InlB
  • Differences in human and animal receptors lead to differential infectivity and ability of Listeria to bind and invade

Trigger Mechanism - Shigella flexneri

  • Shigella flexneri is a Gram-negative, waterborne pathogen that causes bacillary dysentery
  • Invasion requires a type 3 secretion system (T3SS)
  • T3SS secreted proteins directly assist invasion and target the cytoskeleton and host cell GTPases that regulate actin

Trigger Mechanism - Salmonella

  • Salmonella is an enteric pathogen that causes diarrhea
  • SPI-1 invasion is encoded on Salmonella pathogenicity island 1 and encodes a type 3 secretion system
  • T3SS effectors induce membrane ruffling, facilitating internalization

Intracellular Pathogens

  • Intracellular pathogens reside inside host cells to avoid the host immune response and gain access to nutrients
  • Advantages of intracellular residence include a privileged environment, inaccessibility to attack by complement and antibodies, and ready access to nutrients
  • Disadvantages include a hostile environment if the pathogen is not adapted, immune signaling from infected cells leading to apoptosis, and the need to adapt to intracellular survival

Strategies for Intracellular Survival

  • Enter and multiply in non-phagocytic cells using induced endocytosis or macropinocytosis
  • Enter and multiply in phagocytic cells using phagocytosis
  • Three pathways for intracellular survival: intralysosomal, intravacuolar, and cytosolic

Cytosolic Pathogens - Listeria and Shigella

  • Invade cells and escape the vacuole using pore-forming toxins
  • Recruit host cell cytoskeletal proteins and induce actin polymerization, forming actin tails that propel bacteria into adjacent cells

Actin Tails - Promoting Cell-Cell Spread

  • Actin tails promote bacterial spread within organs and tissues
  • Drive bacteria into protrusions that are engulfed by neighboring cells, allowing bacteria to avoid host immunity

Intravacuolar Pathogens

  • Establish intracellular niches by constructing membrane-encompassed compartments
  • Protect cells from cytoplasmic immune sensing molecules
  • Must maintain the integrity of the membrane compartment to survive

Legionella pneumophila - Life Cycle

  • Legionella pneumophila is a Gram-negative, aerobic rod that lives in amoebae in the environment and human macrophages during infection
  • Two lifecycle phases: replicative and infectious/transmissive
  • The infectious phase involves flagellated forms that enter host cells and secrete effectors to prevent lysosome fusion and recruit vesicles from the ER and mitochondria

Legionella pneumophila - Vesicular Manipulation

  • Effectors manipulate vesicle trafficking and LCV surface molecules
  • Prevents LCV-lysosome fusion and recruits vesicles from the ER and mitochondria to make the LCV resemble the Golgi apparatus

Lysis of Host Cells and Metal Ion Acquisition

  • Bacterial pathogens can acquire metal ions through four mechanisms: lysis of host cells, direct acquisition of host proteins, secreted molecules, and direct acquisition of metal ions.

Secreted Molecules (Siderophores and Metallophores)

  • Secreted molecules, such as siderophores and metallophores, are released into the extracellular environment to directly compete with host proteins for metal ions.
  • Siderophores are low-molecular-weight compounds (500-1500 Daltons) that are primarily synthesized by non-ribosomal peptide synthetases (NRPSs) and are regulated by intracellular Fe abundance.
  • Examples of siderophores include hydroxamates and catecholates, which have a high formation constant (Kf) for Fe(III).
  • Metallophores, such as nicotianamine derivatives, have broad specificity for divalent cations and are not regulated by cellular Fe abundance.

The Siderophore Arms Race

  • The siderophore arms race refers to the competition between bacterial siderophores and host siderocalins (e.g., lipocalin-2) for metal ions.
  • Bacteria have evolved to modify their siderophores to evade host siderocalins, such as the synthesis of glycosylated derivatives (e.g., salmochelin).

Direct Acquisition of Metal Ions

  • Bacteria can acquire metal ions directly from the host environment using high-affinity metal ion transporters, such as ATP-binding cassette (ABC) transporters.
  • These transporters consist of an integral membrane channel and a substrate binding protein, which selectively acquire metal ions and exclude chemically similar metals.

Pneumococcal Surface Adhesin A (PsaA)

  • PsaA is a critical virulence determinant in Streptococcus pneumoniae that binds manganese (Mn) with high affinity.
  • The PsaA protein is part of the PsaBCA ABC transporter system, which is essential for virulence.
  • The crystal structures of PsaA bound to Mn and Zn are largely superimposable, but PsaA achieves selectivity for Mn over Zn through reversible binding.

Therapeutic Targets

  • Metal acquisition pathways are crucial for bacterial viability and can be targeted for therapeutic interventions.
  • Targeting zinc uptake pathways (e.g., ZnuD) and manganese uptake pathways (e.g., PsaA) has shown modest efficacy in reducing bacterial burden during infection.

Summary

  • Metal ions are essential micronutrients for all forms of life, and metal acquisition pathways are crucial for bacterial viability.
  • Competition for metal ions at the host-pathogen interface can determine disease outcome, and targeting metal acquisition pathways can be an effective therapeutic strategy.

Evasion of the Innate Immune System by Bacterial Pathogens

  • Bacterial pathogens use various mechanisms to evade the innate immune system, including:
    • Expressing a capsule to prevent phagocytosis and complement binding
    • Mimicking the host through molecular mimicry
    • Producing biofilms to avoid immune detection
    • Evasive tactics to survive phagocytosis
  • Capsules prevent complement binding and phagocytosis of bacterial pathogens by:
    • Preventing C3b binding on the surface
    • Reducing C3 convertase formation
    • Decreasing C5b production and membrane attack complex formation

Steps of Pathogenesis

  • To cause disease, bacterial pathogens must:
    1. Enter the body
    2. Colonize the host
    3. Evade host defenses
    4. Multiply and disseminate
    5. Cause damage to the host
  • Immune evasion is a crucial step in pathogenesis

Kinetics of Innate and Adaptive Immune Responses

  • Bacterial pathogens need to evade immunity upon entry into the host
  • Innate immune responses are immediate, while adaptive immune responses are delayed
  • Bacterial pathogens aim to avoid detection and clearance by the immune system

Evasion of Adaptive Immune Responses

  • Bacterial pathogens can modify key virulence factors to evade adaptive immunity
  • Examples of evasion strategies include:
    • Phase variation in Salmonella to switch between flagella genes
    • Antigenic variation in Neisseria gonorrhoeae to change pili antigens

Phase Variation

  • Phase variation refers to changes in the expression of important virulence proteins that occur at high frequency
  • It allows bacteria to avoid elimination by the host's immune system
  • Examples include:
    • Salmonella switching between flagella genes to avoid immune clearance
    • Neisseria gonorrhoeae altering pili antigens through recombination

Antigenic Variation

  • Antigenic variation involves changing surface antigens through gene shuffling events
  • This allows pathogens to avoid host antibody responses by providing a new antigenic variant
  • Examples include:
    • Neisseria gonorrhoeae using antigenic variation to change pili antigens
    • Other bacterial pathogens using similar mechanisms to evade adaptive immunity

Identifying Virulence Factors

  • Two recognized mechanisms for bacteria to invade host cells
  • One mechanism involves disruption of some Dot/Icm effectors, associated with phenotypes
  • Similar studies: Martinez et al., PLoS Pathog, 2014 (cig2::Tn), and Chao et al, 2016, Nat Rev Micro (TnSeq results)

TnSeq Advantages

  • Capacity to test very large numbers of mutants at once, statistically very powerful
  • Significant reduction in the number of animals used in animal models

Koch's Postulates

    1. The microbe must be found in all organisms suffering from the disease and not in healthy organisms
    1. The microbe must be isolated from the diseased organism and grown as a pure culture
    1. A pure culture of the microbe, when inoculated into a susceptible and healthy host, must reproduce the disease
    1. The microbe must be reisolated in pure culture from the experimentally infected host

Limitations of Koch's Postulates

  • Not applicable in the molecular era

Molecular Era Postulates

    1. The phenotype or property under investigation should be associated with pathogenic members of a genus or pathogenic strains of a species
    1. Specific inactivation of the gene(s) associated with the suspected virulence trait should lead to a measurable loss in pathogenicity or virulence
    1. Reversion or allelic replacement of the mutated gene should lead to restoration of pathogenicity

Identifying Virulence Genes

  • Phenotype: understanding disruption of some Dot/Icm effectors
  • Model: Transformation and antibiotic selection
  • Method: Array into 96-well tray, results from Carey and Newton, PLoS Pathog, 2011

Screening for Virulence Factors

  • Current approach: transformation and antibiotic selection
  • Limitations: labor-intensive and time-consuming
  • Potential improvements: TnSeq, which can test very large numbers of mutants at once, statistically very powerful

Types of Vaccines

  • Live attenuated vaccines: weakened or inactivated pathogens that induce similar immune responses to natural infection
  • Killed whole organism vaccines: inactivated pathogens that stimulate an immune response
  • Toxoid vaccines: detoxified toxins from pathogenic bacteria that neutralize disease symptoms
  • Subunit vaccines: use purified protein, recombinant protein, polysaccharide, or peptide from pathogens
  • Virus-like particle vaccines: resemble viruses but lack genetic material
  • Outer membrane vesicle vaccines: use Gram-negative bacterial outer membrane antigens
  • Conjugate vaccines: combine carrier protein with polysaccharide from pathogens
  • Viral vectored vaccines: use viral vector to deliver pathogen genes
  • Nucleic acid vaccines: use DNA or RNA to stimulate an immune response

Live Attenuated Vaccines

  • Show limited replication in vivo but induce strong immune responses
  • Resemble natural infection, delivering antigens with infection-specific PAMPs
  • One of the most effective methods for generating T cell-based immunity
  • Common methods for attenuation include:
    • Using closely related species causing milder disease
    • Long-term cultivation or passaging
    • Chemically induced multi-site mutagenesis

Toxoid Vaccines

  • Neutralize toxins produced by pathogenic bacteria
  • Detoxified toxins are highly immunogenic but require multiple boosters over lifetime
  • Two licensed toxoid vaccines to date: Tetanus toxin (TT) and Diphtheria toxin (DT)
  • Main risks and weaknesses include producing toxin and positive selection

Antibiotics Overview

  • Antibiotics are molecules that kill or stop the growth of microorganisms, including bacteria, fungi, or viruses.
  • Antibiotics can be derived from nature (natural products), synthesized (synthetic chemistry), or semi-synthetic (modified natural products).

Sources of Antibiotics

  • Bacteria/Fungi: Actinomycetes (e.g., Streptomyces) produce antibiotics, primarily tetracycline and methicillin.
  • Semi-synthetic antibiotics are made by modifying natural products in a lab.

Classifying Antibiotics

  • Antibiotics can be classified as bactericidal (kill bacteria) or bacteriostatic (inhibit growth).
  • They can also be classified as broad-spectrum (effective against both Gram-positive and Gram-negative bacteria) or narrow-spectrum (effective against either Gram-positive or Gram-negative bacteria).

Ideal Antibiotic Properties

  • Irreversible binding to a bacteria-specific target, resulting in bacterial cell death.
  • Ability to penetrate Gram-negative and Gram-positive cell walls.
  • Activity at very low concentrations.
  • No drug-drug interactions.
  • Broad spectrum.
  • High oral availability.
  • Good tissue penetration.
  • Limited ability for resistance to develop.

Antibiotics Targeting the Cell Wall

  • Antibiotics can target the bacterial cell wall, specifically peptidoglycan.
  • Peptidoglycan is composed of N-acetylmuramic acid (NAM) and N-acetylglucosamine (NAG) linked by oligopeptide chains.

Beta-Lactam Antibiotics

  • Beta-lactam antibiotics, such as penicillin, inhibit bacterial cell wall synthesis.
  • Penicillin inhibits DD-transpeptidase (Penicillin Binding Protein, PBP).
  • Vancomycin binds to D-Ala-D-Ala of the stem peptide, inhibiting peptidoglycan synthesis.

Penicillin vs. Vancomycin

  • Penicillin: Derived from fungi (Penicillium chrysogenum), discovered by Alexander Fleming in 1928.
  • Vancomycin: Derived from Actinobacteria (Amycolatopsis orientalis), discovered through soil screening in 1954.

Antibiotics Targeting the Ribosome

  • Antibiotics can target the bacterial ribosome, specifically the 30S and 50S subunits.

Tetracyclines

  • Tetracyclines, such as tetracycline and doxycycline, inhibit protein synthesis by binding to the 30S ribosomal subunit.
  • They prevent amino-acyl tRNA binding to the A site.

Linezolid

  • Linezolid, an oxazolidinone, inhibits protein synthesis by binding to the 50S ribosomal subunit.
  • It blocks the P-site and stops initiation of protein biosynthesis.

Tetracyclines vs. Linezolid: Resistance

  • Tetracycline resistance is often due to ribosomal methylation or efflux.
  • Linezolid resistance is rare, likely due to its novel mechanism of action.

The Problem of Antibiotic Resistance

  • Antibiotic resistance is increasing, and it is a WHO priority.
  • Priority 1 pathogens include Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacteriaceae, which are critical to address.
  • Priority 2 pathogens include Enterococcus faecium, Staphylococcus aureus, and Helicobacter pylori, which are considered high priority.

Definition of Antibiotic Resistance

  • Antibiotic resistance refers to an organism's ability to grow in the presence of an antibiotic designed to kill or inhibit it.
  • Resistance can be caused by various factors, including intrinsic and acquired mechanisms.

Importance of Antibiotic Resistance

  • Antibiotic resistance leads to untreatable infections, longer hospital stays, greater costs, and increased mortality.
  • Several infections are completely untreatable due to antibiotic resistance.

Genetic Basis of Resistance

  • Intrinsic resistance can arise from various mechanisms, including the cell type, lack of target or mutated target, and chromosomal resistance genes.
  • Acquired resistance can occur through horizontal gene transfer, involving extrachromosomal DNA, plasmids, transposons, and phage.

Origins of Resistance

  • Antibiotic producers must protect themselves from their products, leading to the evolution of resistance genes.
  • Many antibiotic producers are resistant to multiple antibiotics, suggesting that these genes are ancient.
  • The origins of resistance can be traced back to environmental bacteria, with evidence from genomics and permafrost sequencing.

Resistance Mechanisms

  • Efflux pumps actively pump antibiotics out of the cell, and many targets are inside the cytoplasm or inner membrane.
  • There are five major families of efflux pumps, with different substrate specificities.
  • Inactivation mechanisms involve the destruction of antibiotics, such as β-lactamases, which cleave the β-lactam ring.
  • Target modification mechanisms involve the modification of the antibiotic target, such as methicillin resistance in Staphylococcus aureus.

Identification of Resistance

  • Antibiotic susceptibility testing is used to determine the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of antibiotics.
  • Methods for testing include disk diffusion, E-test, VITEK, and broth microdilution.
  • Genomics can also be used to detect resistance markers.

Why Do We Have a Resistance Problem?

  • The introduction of an antibiotic is always followed by the emergence of resistance.
  • The antibiotic discovery timeline has slowed, with no new antibiotic classes since the 1990s.

What Can We Do About Resistance?

  • Antibiotic stewardship involves using fewer antibiotics, only when necessary, and stopping unnecessary use in agriculture.
  • Alternative treatments, such as phage therapy, can be used to combat antibiotic resistance.
  • New antibiotics can be discovered through natural product screening, whole cell assays, and target-based screening.

Antibiotic Discovery

  • Natural products, such as those from Streptomyces, are a rich source of antibiotics, but it is challenging to identify new structures and access complex gene clusters.
  • New methods, such as iChips, induction, and clone and express genes, can be used to overcome these challenges.
  • Antibiotic discovery from nature is an excellent source, but it is challenging to identify new structures and access complex gene clusters.

ESKAPE Pathogens

  • ESKAPE pathogens are a group of bacteria that are highly resistant to antibiotics and cause serious infections in healthcare settings.
  • The acronym ESKAPE stands for:
    • E: Enterococcus faecium
    • S: Staphylococcus aureus
    • K: Klebsiella pneumoniae
    • A: Acinetobacter baumannii
    • P: Pseudomonas aeruginosa
    • E: Enterobacter spp.

Staphylococcus aureus

  • S. aureus is a gram-positive, cocci-shaped bacterium that can cause a range of acute and invasive infections.
  • Its genome is enriched with mobile genetic elements, which allows for the transmission of virulence factors, including antibiotic resistance.
  • Methicillin resistance in S. aureus arises from the mecA gene, which is carried on the mobile genetic element SCCmec.

Methicillin-Resistant S. aureus (MRSA)

  • MRSA emerged in 1960, shortly after the introduction of methicillin.
  • MRSA remained rare until the 1980s, but its prevalence has increased significantly since then.
  • The mecA gene confers resistance to almost all β-lactam antibiotics and is transmitted on mobile genetic elements.

Enterococcus faecium

  • E. faecium is a gram-positive, cocci-shaped bacterium that is a commensal organism in the gastrointestinal tract of humans and animals.
  • However, it can also cause healthcare-associated infections (HAIs) and has acquired tailored gene repertoires not present in commensal organisms.
  • The genome of E. faecium is highly dynamic, with a prominent feature being the constant flux of accessory genes through horizontal gene transfer events.

Vancomycin Resistance

  • Vancomycin resistance in E. faecium is carried on mobile genetic elements, which can be transmitted to other bacteria.
  • The vanB operon on Tn1549 is mobilized via horizontal gene transfer within the gastrointestinal tract to E. faecium.
  • De novo evolution of vanB-mediated resistance has also been observed in patients.

WHO Priority Pathogens

  • The World Health Organization (WHO) has classified pathogens into three priority categories for the development of new antibiotic therapeutics:
    • Critical Priority: Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacteriaceae.
    • High Priority: Enterococcus faecium, Staphylococcus aureus, and Klebsiella pneumoniae.
    • Medium Priority: Streptococcus pneumoniae, Haemophilus influenzae, and Shigella spp.

Acinetobacter baumannii and Pseudomonas aeruginosa

  • Two Gram-negative bacterial pathogens that impact human health
  • Both are opportunistic pathogens and major causes of nosocomial infections

Acinetobacter baumannii

  • Gram-negative, short rod-shaped bacterium
  • Motile, but lacks flagella; uses Type IV pili or secreted exopolysaccharide
  • Strict aerobe
  • Classified in the Gamma-proteobacteria
  • Evolved from a low-virulence commensal bacterium to a successful opportunistic pathogen
  • Environmental reservoir is unclear, with some studies suggesting it is ubiquitous in the environment, while others argue it is solely present in hospital environments
  • Relatively large bacterial genome, averaging 3.5 Mbp, with a core genome of only 1,344 genes
  • Pan-genome contains large amounts of non-conserved material, indicating horizontal gene transfer
  • Clinically significant opportunistic pathogen, causing pneumonia, UTIs, wound infections, meningitis, and bacteremia, particularly in ICUs and aged-care facilities
  • Treatment options are limited, with increasing carbapenem-resistance, and phage-therapy showing some success

Pseudomonas aeruginosa

  • Gram-negative, encapsulated, rod-shaped bacterium
  • Unipolar motility, using flagella and Type IV pili
  • Classified in the Gamma-proteobacteria
  • Secretes pigments, including pyocyanin, pyoverdine, pyorubin, and pyomelanin
  • Ubiquitous environmental bacterium, present in soil, water, skin flora, and man-made environments
  • Large bacterial genome, averaging 6.2 Mbp, with a core genome of only 665 genes
  • Horizontal gene transfer accounts for much of the pan-genome
  • Facultative anaerobe, able to grow in microaerophilic or anaerobic conditions
  • Clinically significant opportunistic pathogen, particularly in immunocompromised individuals, causing respiratory tract, urinary tract, and blood infections
  • Frequent colonizer of aseptic medical devices, with high mortality rates associated with pneumonia and bacteremia

Cystic Fibrosis and Pseudomonas aeruginosa

  • P. aeruginosa is a leading cause of morbidity and mortality in cystic fibrosis patients
  • The bacterium rapidly adapts to the mucus in the lungs of CF patients, leading to chronic infection
  • Strains isolated from CF lungs have dramatic genetic, morphological, and physiological changes, rarely seen in non-CF individuals
  • Characteristics of CF clinical P. aeruginosa isolates include mucoidy, lack of virulence factor expression, hyper-mutability, and resistance to multiple classes of antibiotics

Antibiotic Resistance

  • A. baumannii and P. aeruginosa have low antibiotic susceptibility due to multifactorial resistance mechanisms
  • Mechanisms include multidrug efflux pumps, antibiotic-degrading and -inactivating enzymes, low permeability of cellular membranes, and rapid acquisition of new resistance determinants
  • Resistance to common first-line antibiotics, including β-lactams, carbapenems, polymyxins, and aminoglycosides, limits treatment options
  • Decreasing pipeline of new antibiotics and increasing antibiotic resistance is a major concern

Resistance-Nodulation-Division (RND) Family

  • Ubiquitous in bacteria, archaea, and eukaryotes
  • Efflux pumps that are highly prevalent in many Gram-negative bacterial species
  • Function as proton/substrate antiporters, with 7-8 phylogenetic families
  • RND systems involved in drug efflux are AdeABC, AdeFGH, and AdeIJK in A. baumannii, and 13 RND transport systems in P. aeruginosa

MST Review

  • The overall score of the class is 29/35 (85%) with a standard deviation of 3.5 (10.1%) and a median score of 31 (88.6%).
  • 100% of the class passed, with ~3/4 scoring above 80%.
  • Questions that were answered well include diagnostic tests, Koch's postulates, and vaccines.
  • Questions that were not answered well include "choose the incorrect answer" type questions, and some questions were left unanswered.
  • Tips for short answer questions: read the question, avoid "brain dumping", and provide clear explanations with factual backup.

MST 2

  • The MST 2 test will take place on Friday, 3rd May at 10am, lasting 45 minutes and worth 20% of the final mark.
  • The test will consist of short and extended answer questions, covering lectures 14-26, with no multiple-choice questions.

Hospital Acquired Infections

  • Colonization vs infection: understanding the difference is crucial.
  • Study methods used to track hospital acquired infections include surveillance and tracing.
  • Limitations of studies on hospital acquired infections include differences in surveillance methods, regional differences in data sources, and limited evidence of clonal relationships between colonizing and infecting strains.

Paper 1

  • The study investigated the difference between colonization and infection.
  • The study found various results, but limitations included differences in surveillance methods and regional differences in data sources.

Paper 2

  • Multidrug-resistant S. epidermidis is a potential problem in hospitals.
  • The mechanism of resistance, especially to vancomycin, in S. epidermidis is important to understand.
  • Clonal expansion of MDR S. epidermidis in hospitals is a concern.

Scenario

  • To determine the origins, risk factors, and potential outbreak of a hospital pathogen, track the spread of the organism within the hospital.
  • Methods to track the spread include:
    • Determining if the organism has a hospital or community origin.
    • Identifying factors involved in spreading the organism within the hospital.

Bacterial Toxins

  • Bacterial toxins are toxic substances produced and released by bacteria to target other bacterial or host cells.
  • Can be classified into two main types: endotoxins and exotoxins.

Endotoxins

  • Lipopolysaccharides associated with the cell wall of Gram-negative bacteria.
  • Integral part of Gram-negative bacterial cell wall, mainly released when bacteria are lysed.
  • Consists of three parts: O antigen, core, and Lipid A.
  • Lipid A is responsible for toxicity, and TLR4 is the LPS receptor.
  • Pyrogenic, less potent than exotoxins, and can lead to fever, diarrhea, and septic shock.

Mechanism of Endotoxin

  • Endotoxin activates macrophages, which produce IL-1 and TNF.
  • Leads to the activation of complement and the coagulation cascade.
  • Results in hypotension, fever, and inflammation.

Effects of Endotoxin

  • Fever caused by the production of IL-1 by macrophages.
  • Leukopenia followed by leukocytosis.
  • Hypotension, an early marker of Gram-negative bacteremia.
  • Septic shock, which can lead to organ failure and death.

Exotoxins

  • Produced by Gram-positive and Gram-negative bacteria.
  • Highly potent, and avirulent strains have often lost the ability to make toxins.
  • Generally released or secreted from the bacterial cell.
  • Most have a specific mode of action, and can be classified according to their activity.

Exotoxin Classification

  • Classified according to their host cell tropism (e.g., cytotoxin, haemolysin, leukotoxin).
  • Classified according to their site of action (e.g., neurotoxin, cardiotoxin, enterotoxin).
  • Classified according to the disease or bacteria (e.g., diphtheria toxin, anthrax toxin).
  • Classified according to their enzymatic activity (e.g., mono glucosyltransferase, ADP-ribosyltransferase).

A-B Toxins

  • Consist of two components: one component (subunit A) is responsible for the activity, and one component (subunit B) is responsible for binding.
  • Generally, the "A" subunit is not active until it is released into the cell by subunit "B".
  • Can be single-chained or multi-chained.

Examples of A-B Toxins

  • Diphtheria toxin: ADP-ribosylation.
  • Exotoxin A: ADP-ribosylation.
  • Botulinum toxin: Zn2+ protease.
  • Tetanus toxin: Zn2+ protease.
  • Cholera toxin: ADP-ribosylation.
  • Shiga toxin: cleaves 23S rRNA.
  • Anthrax toxin LF: Zn2+ protease.
  • Anthrax toxin EF: adenylate cyclase.
  • CDT binary toxin: ADP-ribosylation.

Diphtheria Toxin

  • Produced by Corynebacterium diphtheria.
  • Causative agent of diphtheria.
  • Encoded by a prophage.
  • First identified toxin, discovered in 1888 by Émile Roux and Alexandre Yersin.
  • Lethal dose in humans is 100 ng/kg.
  • Causes inflammation of the heart muscle and nerves.

ADP-Ribosyltransferases

  • ADP-ribosyltransferase is a common activity of exotoxins.
  • Transfer of an ADP-ribose moiety from NAD+ to target proteins of intoxicated eukaryotic cells.
  • Inhibits the function of target proteins.

Examples of ADP-Ribosyltransferases

  • Diphtheria toxin: inhibits protein synthesis.
  • Pseudomonas exotoxin A: inhibits protein synthesis.
  • Cholera toxin: activates adenylate cyclase.
  • TEC heat-labile toxin: activates adenylate cyclase.
  • Pertussis toxin: activates adenylate cyclase.
  • C.botulinum C2 toxin: inhibits actin polymerization.
  • S.aureus EDIN toxin: disassembles Golgi.
  • CDT binary toxin: inhibits actin polymerization.

Anthrax Toxin

  • Produced by Bacillus anthracis.
  • Causative agent of anthrax.
  • Encoded by a plasmid.
  • Anthrax toxin is a multi-chain A-B exotoxin.
  • Made up of three proteins: Protective Antigen (PA), Edema Factor (EF), and Lethal Factor (LF).

Mechanism of Anthrax Toxin

  • EF converts cellular ATP to cAMP, leading to an increase in fluid.
  • LF inactivates MAPKK, leading to apoptosis and death.

Not all Exotoxins are Proteins

  • Recently, several non-protein toxins have been identified.
  • Not all exotoxins are proteins.

Clostridial Diseases

  • The genus Clostridium is diverse, consisting of Gram-positive, obligate anaerobes that form heat-resistant endospores.
  • While not all Clostridium species are pathogenic, several are important human and animal pathogens.
  • Pathogenic Clostridia can be classified into three categories: neurotoxic, enterotoxic, and histotoxic.

Neurotoxic Clostridia

  • Clostridium tetani causes tetanus.
  • Clostridium botulinum causes botulism, a disease that targets the nervous system.
  • Botulinum toxin (BoNT) is the most potent biological toxin known, causing flaccid paralysis and potentially death.

Enterotoxic Clostridia

  • Clostridiodes difficile (C. diff) is a leading cause of infectious diarrhea in hospitals worldwide.
  • C. diff spores are essential for transmission and persistence in hospitals.
  • C. diff infections can cause a spectrum of diseases, including self-limiting diarrhea, pseudomembranous colitis, toxic megacolon, sepsis, and death.

Virulence Factors of C. diff

  • The major virulence factors of C. diff are two toxins: TcdA (Toxin A) and TcdB (Toxin B).
  • Both toxins are members of the large clostridial toxin (LCT) family.
  • TcdA and TcdB are encoded in the Pathogenicity Locus (PaLoc).
  • TcdA and TcdB are monoglucosyltransferases that glucosylate Rho family GTPases, disrupting the actin cytoskeleton and causing cell death.

Clostridium perfringens

  • C. perfringens is an important pathogen of humans and animals, causing diseases such as clostridial myonecrosis (gas gangrene) and food poisoning.
  • Historically, C. perfringens was typed according to the toxin profile of infecting strains (A-E).
  • C. perfringens produces up to 17 different toxins, including α-toxin, β-toxin, and ε-toxin.
  • α-toxin is essential for gas gangrene, and its mechanism of action involves phospholipase C activity, cleaving cell membrane phosphatidylcholine to phosphorylcholine and diacylglycerol.

C. perfringens Food Poisoning

  • C. perfringens is a major cause of food poisoning globally, with symptoms including abdominal cramping, diarrhea, and vomiting.
  • Ingestion of C. perfringens enterotoxin (CPE) producing strains in under-cooked food can cause food poisoning.
  • CPE is a pore-forming toxin of the aerolysin family, binding to claudin receptors and inducing oligomerization and pore formation, leading to tight junction damage and disruption of epithelial permeability barrier.

Chlamydia

  • Chlamydia is the most common STI in the world, with over 200,000 cases in the UK in 2017.
  • Caused by Chlamydia trachomatis, an exclusive human pathogen.
  • Asymptomatic in 80% of individuals, but can lead to blocked fallopian tubes and infertility.
  • Transmitted via sexual contact and vertical transmission.
  • Recognized as a specific STI in the 1970s, with little to no history before this time.

Chlamydia Disease Symptoms

  • In females:
    • Cervicitis, urethritis, pelvic inflammatory disease, perihepatitis, or proctitis.
    • Untreated, increases risk of infertility and ectopic pregnancy.
    • Infants born vaginally to mothers with genital Chlamydia trachomatis may develop conjunctivitis and/or pneumonia.
  • In males:
    • Urethritis, epididymitis, prostatitis, proctitis, or reactive arthritis.
  • In both sexes:
    • Conjunctivitis, pharyngitis, and lymphogranuloma venereum (enlarged lymph nodes).

Chlamydia Trachomatis Serovars

  • 18 serovars that correlate with multiple medical conditions:
    • Serovars A, B, Ba, and C: ocular infection → chronic conjunctivitis, can cause blindness.
    • Serovars D-K: Genital tract infections, neonatal infections.
    • Serovars L1-L3: Lymphogranuloma venereum (LGV).

Chlamydia Trachomatis Life Cycle

  • Two developmental forms:
    • Elementary body (EB): infectious form, taken up by host cells.
    • Reticulate body (RB): differentiates from EB, replicative form inside host cells.
  • EBs:
    • Spherical, ~0.2 μm diameter.
    • Osmotically stable, less permeable than RBs → environmentally resistant.
    • Preloaded with virulence factors.
    • Internalized following epithelial attachment, enter a vacuole, known as an inclusion.
  • RBs:
    • Spherical, ~0.8 μm diameter.
    • Not infectious.
    • Not stable outside of the host cell.
    • Actively replicate inside the host cell.
    • Enriched in proteins involved in nutrient uptake, ATP generation, and translation.

Chlamydia Trachomatis Intracellular Niche

  • The inclusion:
    • Migrates to microtubule organizing centre.
    • Regulates fusion and membrane dynamics.
    • Inhibits lysosome fusion, but promotes fusion with nutrient-rich vesicles.
    • Scavenges nutrients, especially lipids.
    • Interacts with other organelles, such as lipid droplets, peroxisomes, and mitochondria.

Chlamydia Trachomatis Modifying the Host Response

  • Premature host-cell death limits replication.
  • Chlamydia:
    • Activate pro-survival pathways and inhibit apoptotic pathways.
    • Slow progression of the host cell cycle.
    • Dampen DNA damage response.
    • Induce inflammatory response, but block NF-kB activation.
    • Modify host transcriptome and proteome.

Chlamydia Trachomatis Genomics

  • First genome finished by Sanger sequencing in 1998.
  • Very small genome: 1.04 Mb + 7.4 kb plasmid.
  • Typical of reductive evolution, intracellular lifestyle, and parasitism.
  • Many "essential" bacterial genes missing.
  • Little evidence of horizontal gene transfer.
  • High synteny between different serotypes.

Chlamydia Diagnosis

  • PCR test is most sensitive, targeting a region of the C.pallidum genome.
  • Non-synonymous mutations and pseudogenes differentiate strains causing different diseases.

Syphilis

Treponema Pallidum

  • Three subspecies: T.pallidum subsp.pallidum (syphilis), T.p.subsp.endemicum (bejel or endemic syphilis), and T.p.subsp.pertenue (yaws).
  • Genomes of all subspecies are ~1.14 Mbp.
  • Limited metabolic capacity, many genes for transporters.
  • Human-restricted pathogen, with humans as the only reservoir of T.pallidum.

Treponema Pallidum Genomics

  • T.pallidum has dispensed with most biosynthetic machinery.
  • No oxidative phosphorylation.
  • No need for iron or cytochromes.
  • Many transporters used to scavenge molecules from the host.

Diagnosis of Syphilis

  • Detection mainly by serology, but inability to distinguish between subspecies.
  • Methods:
    • Darkfield microscopy.
    • Direct fluorescent antibody staining.
    • Immunohistochemistry.
    • PCR becoming more common.
    • Point of care tests becoming more available.

Syphilis Treatment and Prevention

  • Historically, treated with Salvarsan, the first synthetic antibacterial compound.
  • Now, penicillin is the primary treatment.

GI Infections

  • Gastrointestinal (GI) infections can be caused by bacterial, viral, or parasitic agents, with common symptoms including diarrhea, abdominal pain, vomiting, fever, nausea, and dehydration.
  • Diarrheal diseases are a leading cause of morbidity and mortality, with 1.31 million deaths in 2017, primarily affecting children.
  • The most common causes of diarrheal diseases are Rotavirus, Shigella, and Salmonella.

Shigella

  • Shigella is a pathogenic biotype of E. coli, with four species that are considered serologically defined anaerogenic biotypes of E. coli.
  • Shigella emerged from non-invasive E. coli through convergent evolution around 35,000-270,000 years ago.
  • Despite its genetic similarity to E. coli, Shigella is still referred to by its original taxonomy due to its clinical significance.

Shigellosis: Disease and Transmission

  • Shigellosis has a very low infectious dose (10-100 bacteria) and is acid-stable.
  • The primary symptom of Shigellosis is diarrhea, which can range from watery to bloody (dysentery).
  • Infection occurs in the colon and rectum, with clinical presentations ranging from asymptomatic to invasive.
  • Shigellosis can be transmitted through person-to-person contact, contaminated food or water, and sexually.
  • There were an estimated 188 million cases of Shigellosis in 2017, with approximately one-third occurring in Asia.

GI Infections

  • Gastrointestinal (GI) infections can be caused by bacterial, viral, or parasitic agents, with common symptoms including diarrhea, abdominal pain, vomiting, fever, nausea, and dehydration.
  • Diarrheal diseases are a leading cause of morbidity and mortality, with 1.31 million deaths in 2017, primarily affecting children.
  • The most common causes of diarrheal diseases are Rotavirus, Shigella, and Salmonella.

Shigella

  • Shigella is a pathogenic biotype of E. coli, with four species that are considered serologically defined anaerogenic biotypes of E. coli.
  • Shigella emerged from non-invasive E. coli through convergent evolution around 35,000-270,000 years ago.
  • Despite its genetic similarity to E. coli, Shigella is still referred to by its original taxonomy due to its clinical significance.

Shigellosis: Disease and Transmission

  • Shigellosis has a very low infectious dose (10-100 bacteria) and is acid-stable.
  • The primary symptom of Shigellosis is diarrhea, which can range from watery to bloody (dysentery).
  • Infection occurs in the colon and rectum, with clinical presentations ranging from asymptomatic to invasive.
  • Shigellosis can be transmitted through person-to-person contact, contaminated food or water, and sexually.
  • There were an estimated 188 million cases of Shigellosis in 2017, with approximately one-third occurring in Asia.

Group A Streptococcus Overview

  • Family: Streptococcaceae
  • Genus: Streptococcus (medically important pathogens)
  • Characteristics:
    • Gram-positive cocci
    • Facultative anaerobes
    • Non-motile
    • Host-adapted
    • Commensal of nasopharyngeal tract and skin
    • Opportunistic pathogen
    • Largely extracellular living
  • Transmission: Aerosols and direct contact

Group A Streptococcus Characteristics

  • Classified based on multiple factors
  • Beta haemolytic
  • Differentiated on basis of group carbohydrate (>12 serological forms)
  • Group A carbohydrate: Polyrhamnose backbone + GlcNAc sidechain
  • Catalase negative: Unable to convert H2O2 -> H2O + O2

Streptococcus pyogenes - Why Worry?

  • Top 10 infectious disease agent
  • Causes ~600,000 deaths/year
  • S.pyogenes infections endemic in some areas
  • Antibiotic resistance is a continual emerging problem
  • No vaccine currently available

Streptococcus pyogenes - Infections

  • Bacterial pharyngitis (Strep throat)
  • Puerperal sepsis (Childbed fever)
  • Necrotising fasciitis (Flesh-eating disease)
  • Scarlet fever
  • Infections: 700 million cases/year (superficial disease) and 1.78 million new cases/year (severe GAS disease)
  • Deaths: >500,000 deaths/year

GAS causes Scarlet Fever

  • Toxin-mediated disease primarily affecting young children
  • GAS produces a variety of distinct toxins (superantigens like SpeA, SpeC, etc.)
  • Superantigen expression leads to hyperstimulation of the host immune system

GAS Epidemiology

  • 2011 Hong Kong scarlet fever outbreak
  • Predominant GAS emm types: emm12 (>80%) and emm1 (>10%)
  • Antibiotic resistance: Macrolide (>95%) and tetracycline (>80%) resistant

Genomics in GAS Epidemiology

  • Importance of genomics in understanding bacterial evolution (e.g., outbreaks)
  • Identifying virulence factors/mechanisms of pathogenesis
  • Designing new vaccines and improving vaccine design
  • Developing new antibiotics
  • Developing new molecular diagnostics
  • Designing public health interventions
  • Monitoring pathogen populations' response

Learning from a Single Genome

  • Complete genome sequence of an emm12 GAS from a scarlet fever patient
  • Features: 1,908,100 bp, 1942 CDS, φ1 (46.4 kb), φ2 (45.1 kb), and ICE (64.9 kb)

Learning from Comparative Genomics

  • Comparative analysis of HKU16 (emm12) and published non-scarlet fever emm12 genomes (MGAS9429 and MGAS2096)
  • Identification of new prophages and ICE elements

The Genus Mycobacterium

  • The genus Mycobacterium consists of 190 species, including major human pathogens such as M. tuberculosis and M. leprae.
  • Mycobacteria are aerobic, rod-shaped bacteria that stain acid-fast using the Ziehl-Neelsen stain, and have a variable Gram stain.
  • The cell wall of Mycobacteria is unique, with a thicker cell wall than other bacteria, composed of hydrophobic and waxy mycolic acids, and outer lipids linked to peptidoglycan by arabinogalactan.

Mycobacterial Lipids

  • Mycobacterial lipids make up 30-60% of the cell's dry weight and are important for both structure and virulence.
  • Many lipids are unique to each Mycobacterial species, and can activate or suppress the immune system, help survive inside macrophages, and evade the immune system.

Mycobacterium Tuberculosis

  • M. tuberculosis is the causative agent of human tuberculosis, primarily infecting the lungs, but can disseminate to other tissues.
  • It is a slow-growing, intracellular pathogen that divides every 18-24 hours, and takes 3-4 weeks to form colonies.
  • M. tuberculosis has a complex lipids, including phthiocerol dimycocerosates (PDIMs), phenolic glycolipids (PGLs), and sulfolipids (SLs), which are important for virulence.

M. tuberculosis Virulence Factors

  • PE/PPE family proteins are secreted by type VII secretion systems and comprise nearly 10% of the genomic coding potential.
  • Five type VII secretion systems (ESX-1 to ESX-5) are involved in delayed phagosome maturation, phagosome rupture, and cellular necrosis.

Extrapulmonary TB

  • Extrapulmonary TB occurs when M. tuberculosis disseminates outside of the lungs, and can occur in the pleural cavity, peritoneum, nervous system, genitourinary system, bones, and multiple sites simultaneously.
  • It is more common in immunosuppressed individuals and HIV patients, and accounts for ~20% of all TB cases.

Tuberculosis Epidemiology

  • In 2019, there were 10 million new cases of TB disease, with 1.5 million deaths, and 2 billion people with latent TB.
  • The greatest disease burden is in low- and middle-income countries, with 10% of cases co-infected with HIV.

Origins of M. tuberculosis

  • The origins of M. tuberculosis are still uncertain, but it is thought to have emerged from environmental mycobacteria through a stepwise adaptation to the intracellular environment.
  • The "out of Africa" hypothesis suggests that the oldest relatives of M. tuberculosis are found exclusively in the Horn of Africa, and Africa has the largest diversity of M. tuberculosis lineages.

M. tuberculosis Vaccination

  • The M. bovis BCG vaccine is the most widely used vaccine against TB, but it has highly variable protection and unknown duration of protection.
  • There are 14 vaccine candidates in clinical trials, with the most promising being M72/AS01E, a recombinant fusion protein of two M. tuberculosis antigens, with ~50% efficacy in adult TB-positive populations.

M. tuberculosis Diagnosis and Treatment

  • Microscopy and culture are still the main diagnostic tools for active disease, but new tools such as GeneXpert and line-probe assays are being developed.
  • TB treatment typically involves multiple drugs given simultaneously over a long period, with 2 months of intensive treatment followed by 4 months of continuation treatment.
  • Drug-resistant variants are emerging, with 480,000 cases of multidrug-resistant TB (MDR-TB) and 48,000 cases of extensively drug-resistant TB (XDR-TB) in 2018.

Non-Tuberculous Mycobacteria (NTM)

  • Member of the Actinobacteria, with 190 species within the genus Mycobacterium, including major human pathogens M. tuberculosis and M. leprae.

Mycobacterium leprae

  • Causative agent of Leprosy (Hansen's disease), a chronic infectious disease of nerves, skin, eyes, and nasal mucosa.
  • Transmission is not completely defined, but believed to be spread by coughing or contact with nasal secretions from an infected individual.
  • Epidemiology: >200,000 cases per year in >100 countries, with 80% of new cases in India, Indonesia, and Brazil.
  • Armadillos, squirrels, and non-human primates are reservoirs.

M. leprae Pathogenesis

  • Incubation period: 9 months to 20 years.
  • Multiple forms of disease: tuberculoid (paucibacilliary) and lepromatous (multibacilliary).
  • Tuberculoid: cell-mediated immune response limits M. leprae growth, with multiplication at site of entry and Schwann cell invasion.
  • Lepromatous: impaired cell-mediated immune response, microbes proliferate within macrophages, and extensive penetration of M. leprae to distal sites.
  • Treatment: dapsone + rifampicin + clofazimine for 6-12 months.

M. leprae Growth and Culture

  • Very slow growing, with a doubling time of 12-14 days.
  • Growth at 33°C, with tropism for peripheral nerves (lower temp) and Schwann cells and macrophages.
  • Cannot be grown in the laboratory, and is an intracellular pathogen.
  • Grown for study in 9-banded armadillos and footpads of athymic nude mice.

M. leprae Evolution/Genomics

  • Highly reduced genome (3.3Mb).
  • Opportunistic pathogen, seen in immunocompromised hosts.
  • Increasing incidence over the last 20 years (~16/100,000).
  • Treatment: 12-18 months of antibiotic therapy, with side effects common and treatment cost high.

Mycobacterium abcessus

  • Emerging as a significant cause of NTM pulmonary disease in immunocompromised individuals.
  • Produces biofilms, providing protection inside lungs of COPD/CF patients and protecting from drug treatment.
  • Intracellular pathogen, surviving in macrophages.
  • Causes skin infections, linked to acupuncture, tattooing, liposuction, and cosmetic surgery.
  • Increasingly associated with cystic fibrosis patients, with 13% of CF patients having NTM infection.

Cutaneous NTM Infections

  • Most frequently caused by M. marinum (SG), M. ulcerans (SG), M. chelonae (RG), M. abcessus (RG), and M. fortuitum (RG).
  • High levels of antibiotic resistance, with water as the most common source of infection.

Mycobacterium marinum

  • Causes "fish tank" granulomas, a pathogen of fish and humans, caused by direct contact of skin with contaminated source.
  • Slow growing, with optimal temp of 30-32°C, and closely related to M. ulcerans.

Mycobacterium chelonae

  • Often presents as disseminated disease, with multiple lesions, and can involve bone/joint infections.
  • Mainly seen in immunosuppressed individuals, with high levels of resistance to standard anti-mycobacterial drugs.

Mycobacterium chimaera

  • Associated with infection during heart bypass surgery, with risk of 50% mortality and difficult to treat.
  • Slow growing, with no diagnostic tests, and a median incubation period of 17 months.

Anti-Mycobacterial Drugs

  • Isoniazid: prodrug activated by mycobacterial enzyme KatG, binds tightly to InhA, and blocks fatty acid synthesis and mycolic acid synthesis.
  • Ethambutol: inhibits arabinosyl transferase (embCAB operon), disrupting arabinogalactan biosynthesis and mycobacterial cell walls.

New Anti-Mycobacterial Drugs

  • Bedaquiline: newest anti-TB drug (2012), diarylquinoline family, inhibits mycobacterial ATP synthase, and is bactericidal, active against replicating and dormant organisms.
  • Delaminid/pretomanid: nitroimidazoles, inhibit cell wall synthesis (blocks mycolic acid synthesis), approved for MDR-TB.
  • Telacebec (Q203): recently completed phase 2 (2020) for TB, targets cytochrome bc1, depleting ATP from cells, and highly active against M. tuberculosis.

Streptococcus pneumoniae

  • First discovered in 1881 by Louis Pasteur, later identified by Carl Friedländer & Albert Fränkel (1884)
  • Strictly human-adapted, no environmental reservoir
  • Can reside asymptomatically in the upper respiratory tract
  • Highly successful pathogen, estimated to cause 1.6 million deaths annually, one of the leading causes of mortality globally

Global Burden of Disease

  • 1.6 million deaths annually, one of the leading causes of mortality globally
  • USD$4 billion in costs (healthcare and loss of productivity) in the US alone
  • WHO-designated priority pathogen, leading cause of community-acquired bacterial pneumonia (up to 50%)
  • Ever-increasing rates of antibiotic resistance

S. pneumoniae Features

  • Gram-positive, encapsulated, non-motile diplococcus
  • Size: 0.5-1.5 µm
  • Cell wall composed of:
    • Peptidoglycan
    • N-acetylglucosamine (GlcNac) and N-acetylmuramic acid (MurNac)
    • Teichoic acid (TA) and lipoteichoic acid (LTA)
  • Most strains also encapsulated, capsule determines the serotype

Human Specialist

  • Relatively small genome (2.0-2.1 Mbp), with little over 2000 genes
  • Host-mediated genome decay
  • Nutritionally fastidious, scavenges nutrients from the host wherever possible
  • Auxotrophic for numerous amino acids, scavenges catalase by lysing host RBCs (α-haemolysis)
  • Multiple incomplete biosynthetic pathways, e.g. glutathione
  • Can adopt different 'lifestyles': carriage, disease

Pneumococcal Carriage

  • Carried asymptomatically in the nasopharynx (~60% of children, ~5% of adults)
  • Primary source of host-to-host transmission
  • Carriage without dissemination often associated with specific serotypes
  • Colonization is necessary for disease, followed by dissemination or invasion into deeper tissues

Pneumococcal Invasion

  • Middle ear – otitis media: inflammation and pain in the middle ear, can result in perforated ear drums and deafness
  • Lungs – pneumonia: inflammation of the respiratory tract and lung tissue, results in cough, shortness of breath, and fluid accumulation in lungs
  • Blood – bacteraemia: also called blood poisoning, results in systemic inflammatory response
  • Brain – meningitis: severe infection of the CNS, mortality rates up to 37%, long-term or permanent morbidity in 30-50% of surviving patients

Pneumococcal Pneumonia

  • Responsible for > 1M deaths annually, ~7% of global deaths
  • Greatest risk in the young and old
  • Can be initiated by viral infection, requires antibiotic intervention

How to be a Successful Pathogen

  • Pneumococcal capsule: extracellular polysaccharide that coats the bacterial cell wall
  • Phase variation: random, high-frequency, reversible switching of gene expression
  • Natural competence: ability to take up DNA from the environment
  • Toxins (Pneumolysin): cholesterol-dependent cytolysins, binds to cholesterol on host cells, causing lysis

Capsule – What is it?

  • Extracellular polysaccharide that coats the bacterial cell wall
  • Made up of repeating sugar molecules (saccharides), poorly immunogenic
  • Currently ~100 different serotypes, varying in sugar composition and linkage

Capsule – What does it do?

  • Primary virulence determinant in S. pneumoniae
  • Reduces mucociliary clearance, prevents complement deposition on the cell surface
  • Facilitates survival in the blood, allows escape from NETs, and prevents opsonisation by antibodies

Therapeutics – Vaccines

  • Pneumovax-23 (Merck): polysaccharide vaccine, recommended for adults
  • Prevenar 13 (Pfizer): polysaccharide conjugate vaccine, given to infants
  • Serotype replacement: emergence of new serotypes due to selective pressure of vaccination

Therapeutics – Antibiotics

  • β-lactams (e.g. penicillin or amoxycillin), macrolides, tetracyclines, and cephalosporins are used
  • Resistance reported up to 80% (!) in some countries
  • Multi-drug resistance in up to 46% of isolates

Burkholderia Cenocepacia and Granulomatous Disease

  • B. multivorans, Aspergillus, and Burkholderia are associated with granulomatous disease.
  • B. cenocepacia is disproportionately associated with Cepacia syndrome, a necrotizing pneumonia.

Infections in Cystic Fibrosis Patients

  • Bcc infections are common in cystic fibrosis (CF) patients, affecting approximately 5% of them.
  • These infections are associated with high rates of morbidity and mortality in CF patients, with a higher mortality rate than P. aeruginosa.
  • Bcc infections lead to enhanced rates of tissue damage, impacting lung function and making lung transplants necessary.

Burkholderia Pseudomallei and Melioidosis

  • B. pseudomallei is a major cause of melioidosis, a disease with high mortality rates (up to 40% in some areas).
  • Melioidosis is often underdiagnosed, making it essential to diagnose and start antimicrobial therapy early.
  • B. pseudomallei infections can cause sepsis or localized abscesses in various organs, including the lungs, brain, soft tissue, and kidneys.

Pathogenesis Mechanisms

  • Burkholderia species use various mechanisms to survive within hosts, including hiding PAMPs (pathogen-associated molecular patterns) and blocking the antimicrobial response.
  • They employ secretion systems (type 3/6) to deliver effectors into host cells.
  • Carbohydrates play a crucial role in pathogenesis, contributing to a protective barrier around the bacteria.

Glycosylation and Virulence

  • Glycosylation is essential for virulence in Burkholderia species, with general glycosylation systems targeting multiple protein substrates.
  • Flagellin glycosylation is observed in all pathogenic Burkholderia species, allowing them to evade immune recognition.
  • General glycosylation leads to the installation of a conserved trisaccharide on proteins in the periplasm.

Immunogenic Properties of Glycans

  • Protein glycosylation is required for virulence in Burkholderia species, but it is also highly immunogenic.
  • Humans mount a robust humoral response to the Burkholderia glycan used for general glycosylation, which can be used to detect exposure to Burkholderia species.

Workshop 7: Identifying New Pathogens

Historical Context: 1976

  • July 1976: Top hit record was "Kiss and Say Goodbye" by The Manhattans
  • Football: VFL (not AFL) with no interstate teams, suburban grounds were used, and teams like Fitzroy and South Melbourne existed
  • US President: Gerry Ford

Legionnaires' Disease Outbreak

  • July 1976: Outbreak of a new disease at the State Convention of Legionnaires at the Bellevue-Stratford Hotel in Philadelphia, Pennsylvania, USA
  • 182 cases of a disease spread via the respiratory tract
  • 29 deaths (16% mortality rate)

Disease Identification

  • December 1977: Papers published outlining the disease and the Gram-negative bacterium (Legionella pneumophila) that caused the disease
  • Identification methods used: animal models, antisera, Gram stain

Investigation in 1976

  • No genomics, no PCR, limited mass spectrometry
  • If time-traveling with modern equipment, would need:
    • Equipment to investigate the outbreak
    • Consumables to pack into the Tardis

This quiz covers genomics, microbial evolution, and epidemiology in bacteriology, with suggested readings from Prescott's Microbiology and Bacterial Pathogenesis.

Make Your Own Quizzes and Flashcards

Convert your notes into interactive study material.

Get started for free
Use Quizgecko on...
Browser
Browser