Conjugation Plasmids and DNA Transfer Efficiency Quiz

ScenicBongos avatar
ScenicBongos
·
·
Download

Start Quiz

Study Flashcards

132 Questions

What is the role of pili size in conjugation?

It may influence the efficiency of DNA transfer

Which of the following is true about transposons?

They are segments of DNA that can move within a genome

What happens when both conjugation occurs in a cell?

Both the donor and recipient cells have a complete copy of the plasmid

What is the main function of a conjugative plasmid in conjugation?

To be replicated in the donor and transferred through the pilus

What is the significance of transposons jumping onto a conjugative plasmid?

Results in the acquisition of resistance or virulence genes by transposition

What are some factors that influence recipient cell uptake of exogenous DNA?

The presence of various promoters and terminators

Which process involves the release of DNA in genetic transformation through the active secretion by live cells?

Active secretion

In genetic transformation, what role do plasmids play in the process of donor cell releasing DNA?

Plasmids release small fragments during passive leakage

What is the primary concern associated with the transfer of antibiotic resistance genes between bacterial strains?

Risk of spreading resistant genes to the environment

How does intraspecies transfer primarily occur within a species?

Via sexual reproduction and parasexual processes

What is the main difference between generalized and specialized transduction?

Generalized transduction can transfer any part of the bacterial genome, while specialized transduction transfers specific parts near the prophage integration site.

Which type of transduction involves the transfer of botulinum toxin or cholera toxin genes?

Specialized transduction

What happens when a prophage is excised from the bacterial chromosome in specialized transduction?

The recipient cell receives a specific set of genes from the donor.

In specialized transduction, why are only close genes to the prophage integration site affected?

To ensure the recipient receives a specific set of genes.

Which statement accurately describes how generalized transduction introduces new genes into the recipient?

By transferring any part of the bacterial genome through recombination with the recipient's chromosome.

What distinguishes generalized and specialized transducing phages based on their virulence factor encoding abilities?

Generalized phages do not encode virulence factors, specialized phages do.

What is the primary reason for slipped-strand mispairing leading to insertions or deletions in DNA?

Errors in DNA replication

Which of the following mechanisms can introduce genetic material from closely related species into bacterial genomes?

Natural transformation

What is the consequence of hyper slipped-strand mispairing where a nucleotide is added or subtracted?

Premature stop codon or nonfunctioning protein

Which process involves the exchange of genetic material within the same genome?

Intra-genomic recombination

During chronic infection with 'non-issue causing' colonizing microbes, approximately how much of the genome can be replaced in 40 years?

50%

What is the main result of variable motility due to slip strand mispairing in multiple phase variable genes?

Change in gene regulation

Which process involves bacteria taking up free DNA fragments from the environment?

Natural transformation

What role does active secretion of DNA play in antigenic variation?

Enables bacteria to share genetic material and surface antigen genes

How does natural competency contribute to antigenic variation in bacteria?

Enables bacteria to uptake and incorporate foreign DNA, potentially changing their antigenic profiles

What is the main outcome of the interaction between active DNA secretion and natural competency in bacteria?

Facilitation of rapid antigenic evolution

How does reversible DNA sequence change differ from antigenic variation?

Antigenic variation involves change via DNA recombination

In the context of bacterial evolution, what is the significance of phase variation?

Enables bacteria to switch between only two variants of a surface antigen

What is the specific role of phage receptors in the infection process?

Attaching to the bacterial cell to initiate the infection process

During which stage of phage replication does the genetic material take control of the host cell's machinery?

Biosynthesis

What occurs during the lysis and release phase of phage replication?

Release of phage particles from the host cell

What is the primary function of the phage capsid during the infection process?

Facilitating attachment to the bacterial cell

Why is it essential for phages to redirect the host cell's machinery during replication?

To produce new genetic material for the phage

What is a key requirement of Koch's postulate for confirming that a microbe causes a specific disease?

The microbe must cause the disease symptoms in a lab setting without infecting a host

Who among the 'Founding Fathers of Microbiology' is credited with the pasteurization of milk?

Louis Pasteur

Which of the Founding Fathers of Microbiology is known for being the 'father of microbiology' due to his invention of the first microscope?

Antonie Van Leeuwenhoek

What distinguishes Joseph Lister's contribution among the Founding Fathers of Microbiology?

Discovery of sterilization methods for surgery equipment

Who developed the first vaccine for polio among the Founding Fathers of Microbiology?

Jonas Salk

Why is it important to target unique bacterial components when developing antimicrobial agents?

To ensure that antimicrobials selectively attack bacteria without harming the host's cells

What is the significance of targeting essential functions for bacterial growth with antimicrobial agents?

It effectively kills or inhibits the growth of bacteria

How does targeting virulence factors impact bacteria's ability to cause infections?

It disarms the bacteria, making them less able to establish infections or evade the immune system

Which process do antimicrobials target by focusing on DNA replication, protein synthesis, and cell division?

Critical processes for bacterial survival and proliferation

In developing antimicrobials, why is it crucial to consider the host's cells?

To reduce toxicity and side effects in humans

What is the primary advantage of targeting unique bacterial components over broadly attacking all cellular functions?

It minimizes damage to host cells

How does targeting essential functions for bacterial growth with antimicrobials relate to bacterial survival?

It effectively kills or inhibits the growth of bacteria

What is the primary intention behind targeting virulence factors with antimicrobial agents?

To weaken bacteria's ability to infect host organisms and cause diseases

Which characteristic makes targeting unique components in bacteria more advantageous than generalized antimicrobial approaches?

Reduces the risk of side effects in humans

How does focusing on essential functions for bacterial growth aid in combating bacterial infections?

Eliminates bacteria effectively without harming host cells

What is the primary role of Bacitracin in inhibiting bacterial cell wall synthesis?

Disrupts the synthesis of cell walls

Which biochemical process does Fluoroquinolone impact to exert its effect?

Inhibits DNA gyrase/topoisomerase

What is the main target of Beta-lactams in bacterial cells?

Transpeptidation process

How does Cycloserine exert its inhibitory action on bacteria?

Promotes DNA gyrase activity

Which of the following is a direct impact of microbes requiring folic acid for DNA biosynthesis?

Requirement for folic acid in diet

How do Beta-Lactam antibiotics work?

Block the transpeptidation step in cell wall synthesis

What is the role of D-ala in peptidoglycan synthesis of Gram-negative bacteria?

It is used to link one brick to another during transpeptidation

What sets Gram-positive bacteria apart from Gram-negative bacteria in terms of their cell wall synthesis?

Formation of an interpeptide bridge

What happens when Beta-Lactam antibiotics block the transpeptidation step?

Causes the formation of a weak cell wall

Why are Gram-positive bacteria harder for antibiotics to penetrate compared to Gram-negative bacteria?

They possess an interpeptide bridge in their cell wall

What makes Beta-Lactam antibiotics effective against bacterial infections?

They disrupt the formation of strong chemical bonds between peptidoglycan units

In peptidoglycan synthesis, what is the consequence of inhibiting transpeptidation?

Formation of a thinner cell wall

Which step in cell wall synthesis do Beta-Lactam antibiotics specifically target?

'Cementing' by transpeptidation

What effect does the formation of an interpeptide bridge have on Gram-positive bacteria?

It increases resistance to antibiotics

What is the main consequence of targeting transpeptidation in bacterial cell wall synthesis with Beta-Lactam antibiotics?

Weakening of peptidoglycan structure

What is the mechanism by which bacteria have developed resistance to beta-lactams?

By acquiring beta-lactamases to cleave the beta-lactam ring

What action do bacteria take to counteract the use of beta-lactam antibiotics combined with beta-lactamase inhibitors?

Acquiring alternate transpeptidases like PBPs

How do bacteria make beta-lactam antibiotics ineffective?

By evolving to replace D-ala with other amino acids in cross-linking peptides

What role do penicillin-binding proteins (PBPs) play in bacterial resistance to antibiotics?

They are acquired by bacteria as a defense mechanism

How does vancomycin-resistant Enterococcus (VRE) evade the inhibitory action of vancomycin?

By replacing D-ala-Dala with D-ala-Delactate in cross-linking peptides

What strategy are scientists exploring to combat bacterial resistance to beta-lactams?

Developing analogs that mimic the structure of D-ala

How does cycloserine inhibit bacterial growth?

By preventing L-ala conversion to D-ala by racemase

Why are beta-lactam antibiotics rendered ineffective against bacteria that have acquired beta-lactamases?

The beta-lactam ring is cleaved before transpeptidase can recognize it

How do bacteria make Beta-Lactams ineffective?

They replace D-alanine with other amino acids in cross-linking peptides

What is a common strategy used by bacteria to resist Beta-Lactams?

Utilizing Beta-Lactamases to cleave the Beta-Lactam ring

Why is Mycoplasma resistant to Beta-lactam antibiotics?

It does not have a cell wall

What is a common mechanism of resistance found in Acinetobacter baumannii?

Efflux systems

Which bacterial species is intrinsically resistant to vancomycin due to the termination of its pentapeptides in D-ala-D-lactate?

Leuconostoc

What is the approximate spontaneous mutation rate frequency for antibiotic resistance?

One mutation per 10^8 cells

Which bacterial alteration causes methicillin resistance in Staphylococcus?

Mutation of penicillin-binding proteins

What type of bacterial mutation can cause resistance to quinolones in E. coli?

Mutation of DNA gyrase

What role do altered ribosomes play in bacterial resistance to antibiotics?

Altered structure preventing correct antibiotic binding

How do porin mutations contribute to bacterial antibiotic resistance?

By reducing antibiotic uptake through the cell membrane

What is a specific advantage of phage therapy compared to antibiotics?

Can evolve host range mutants

What is a characteristic of lytic phages used therapeutically in phage therapy?

They replicate rapidly

Why did phage therapies drop after the introduction of antibiotics?

Antibiotics were considered miracle drugs

What is a drawback associated with early phage therapy experiments?

Limited controls in experiments

Why are lytic phages preferred over temperate phages in phage therapy?

They are less pathogenic

What makes phages unique compared to antibiotics in terms of administration?

They can be taken orally

What issue arises due to the large number of unidentified ORFs found in phages?

Difficulty in determining their activity

In what context can bacteriophages be used as biomarkers?

Microbiota drivers investigation

What is the historical significance of phage typing in microbiology?

A method of identifying bacteria based on their susceptibility to specific bacteriophages

What term did Felix d’Herelle coin to describe viruses that infect bacteria?

Bacteriophages

What is the outcome of the destruction of bacterial DNA during the bacteriophage lifecycle?

Lysis of the bacterial cell

What is the primary function of phage display in microbiology?

Creating a library of peptides or proteins displayed on the surface of phages

What is the relationship between bacteriophages and cloning vectors?

Cloning vectors are derived from bacteriophages

What happens when a bacterial cell is lysed during the lytic/virulent phage lifecycle?

The bacterial cell is killed by the phage particles released.

What contributes to the virulence of the bacterium in cases where lysogen-carrying bacteria are present?

Phage genes carried by the bacteria.

How do eukaryotic viruses differ from bacterial viruses in terms of their effect on host cells?

Eukaryotic viruses can cause chronic infections like HIV in host cells.

Which scenario can result from an Eukaryotic virus mutating to change host preferences?

Eukaryotic virus becoming resistant to immune attacks.

In what way do temperate phages differ from lytic/virulent phages in their impact on infected cells?

Lytic/virulent phages can contribute to increased virulence of infected cells.

What allows C. Difficile to survive passage through the stomach when ingested?

Formation of spores

Which factor is directly linked to the risk of C. Difficile colonization in healthcare settings?

Length of hospital stays

What is the primary role of Toxin B in causing pseudomembranous colitis symptoms?

Causing inflammation in the colon

In the mechanism of action of Toxin A and Toxin B, what type of proteins do they inactivate by glucosylation?

G proteins

Which characteristic of C. Difficile contributes to its persistence in the environment for many months?

Formation of spores

How can members of a hospital transmit C. Difficile spores to other patients?

Through the air as aerosols

Which condition is ideal for C. Difficile to overgrow in the colon?

Use of probiotics

Which characteristic of C. Difficile makes it a challenge to eradicate from healthcare settings?

Formation of biofilms on surfaces

What is the primary role of IgG response to ToxA when faced with toxigenic c.diff?

Induces symptomatic CDI

How does the use of antibiotics contribute to the proliferation of C.diff?

Removal of beneficial bacteria

What consequence arises when an individual acquires Toxigenic c.diff WITHOUT IgG response to ToxA?

Symptomatic CDI

What happens when Non-toxigenic c.diff is acquired?

Asymptomatic colonization

Why is CDI more prevalent in hospitals compared to outside settings?

Opportunistic after antibiotic use

What is the impact of antibiotics on the bacteria that aid in defense during an infection?

Removing them from the gut microbiota

What determines whether an individual with Toxigenic c.diff will be symptomatic or asymptomatic?

Composition of microbiota

What is the significance of the release of toxins causing organ failure?

Inducing systematic inflammation

How does the acquisition of Non-toxigenic c.diff differ from acquiring Toxigenic c.diff WITH IgG response to ToxA?

Asymptomatic colonization vs. symptomatic CDI

Why are vancomycin and metronidazole preferred for treating Clostridium difficile infections?

They are ineffective against normal gut flora

Why is it important to stop treating Clostridium difficile infections with the antibiotics that led to the disease?

The antibiotics can kill off normal gut flora

Why is vancomycin not effective against Bacteroides species?

Vancomycin cannot penetrate the outer membrane of Bacteroides species

Why is metronidazole not effective on some gut bacteria?

Some gut bacteria deactivate metronidazole with enzymes

What makes Clostridia a polyphyletic group?

Their uncertain relationships between species within the group

What characteristic protects Clostridia from environmental stressors?

Endospore formation

What happens if Pseudomembranous Colitis is left untreated?

'Patch coalescing' into septic shock

What is the purpose of Fecal Transplant in dealing with Clostridium difficile (C.Diff) infections?

Introduce healthy flora to the patient

Which Clostridium species is known for causing gas gangrene in open wounds?

Clostridium perfringens

What is the function of Butyrate produced by certain groups of Clostridium bacteria in the colon?

Suppress pathogen proliferation

Which toxin, when released by Clostridium tetani, results in violent muscle spasms?

Tetanus toxin

What is the main benefit of using Clostridium acetobutylicum engineered into E. coli?

Production of short chain alcohols

What is the primary effect of Butyrate on the colon's barrier integrity?

Increase in epithelial cell proliferation

Which Clostridium bacteria was used by Chaim Weizmann to produce short chain alcohols?

Clostridium acetobutylicum

What is the role of Botulinum toxin released by Clostridium botulinum?

Inhibits release of acetylcholine from axons

What is a characteristic of Clostridium bacteria that produce Butyrate?

Ferment non-digestible carbs to form Butyrate

Study Notes

Conjugation Plasmids

  • In conjugation, the pili size might influence the efficiency of DNA transfer
  • Donor carrying plasmid creates a pilus that is attached to the recipient
  • Conjugative plasmid is replicated in the donor and then single-stranded transferred through the pilus to the recipient
  • Recipient can now express plasmid genes
  • Results in the acquisition of resistance or virulence genes by transposition

Genetic Transformation

  • Genetic transformation is a technique used to introduce foreign genetic material into the genome of living organisms
  • Process involves two main components: vector (plasmid carrying a selectable marker gene) and target bacteria or other organism(s)
  • Steps to achieve successful transformation: donor cell releases free circular DNA, recipient cell engulfs the plasmid, and plasmid replicates and integrates into the host genome or remains episomal
  • Antibiotic resistance genes can be transferred between bacteria, and non-antibiotic resistance genes can also be transferred, indicating a broader potential impact on environmental communities

Recipient Cell Uptake

  • Recipient cell uptake of exogenous DNA plays a crucial role in genetic transformation
  • Factors influencing uptake efficiency: size, shape, and chemical modification of DNA molecule, presence of promoters and terminators, and biological properties of recipient cell type

Intraspecies Transfer

  • Intraspecies transfer occurs primarily through sexual reproduction and parasexual processes like illegitimate recombination
  • Interspecies transfer happens through horizontal gene transfer mechanisms like transformation, conjugation, and transduction

Donor Cell Releasing DNA

  • Process of genetic transformation relies on the ability of recipient cells to take up and incorporate foreign DNA
  • Donor cells release DNA through passive leakage, active secretion, or programmed cell death

Transduction

  • Transduction is a mechanism of horizontal gene transfer that occurs through bacteriophages
  • Generalized transduction: phage infects bacterium and begins lytic cycle, can transfer any part of bacterial genome
  • Specialized transduction: occurs with lysogenic phages, transfers specific parts of bacterial genome, and DNA transferred is usually located near the prophage's integration site

Mechanisms of Genetic Diversity

  • Reassortment, diversification, natural transformation, point mutation, intra-genomic recombination, and slipped-strand mispairing are mechanisms that create genetic diversity

Genetic Modification

  • Mutation: DNA sequence changes that are usually non-reversible
  • Reversible DNA sequences: change via DNA recombination or mutations with repeated sequences
  • Antigenic variation: alternate expression of multiple variants of surface antigen
  • Phase variation: on-off expression or switching between only two variants of a surface antigen

Phage Infection and Replication

  • Attachment: phage attaches to a bacterial cell by binding to specific receptor sites
  • Penetration: phage injects its genetic material into the host cell
  • Biosynthesis: phage's genetic material takes over the host cell's machinery
  • Assembly: newly synthesized phage genomes and capsids are assembled into complete phage particles
  • Lysis and Release: phage concludes its replication cycle by lysing the host cell, releasing new phage particles

Microbiology

  • Koch's postulate: microbe must be associated with symptoms of the disease, isolated from the lesion, and reproduce the disease in a susceptible host
  • Features of a good target for antimicrobial agents: targeting unique bacterial components, essential functions for bacterial growth, and virulence factors
  • Antibiotic resistance: targeting cell wall synthesis, ribosome, and DNA gyrase/topoisomerase

Cell Wall Synthesis

  • Synthesis of peptidoglycan: gram-negative bacteria, gram-positive bacteria, and transpeptidation
  • Beta-lactam antibiotics: inhibit transpeptidation, and bacteria develop resistance by acquiring beta-lactamases, alternate transpeptidases, and modifying the terminal amino acid

ESKAPEE Pathogens

  • Enterococcus faecium (VRE)
  • Staphylococcus aureus (MRSA)
  • Klebsiella pneumonia (ESBL-producing)
  • Acinetobacter baumannii
  • Enterobacter species
  • Escherichia coli

Mechanisms of Resistance to Antimicrobials

  • Natural resistance: lack of permeability, lack of target, and chromosomal-encoded efflux pumps

  • Mutation: spontaneous mutation rate frequency, reduced uptake, altered target, and mutation of penicillin-binding proteins

  • Horizontal gene transfer: sharing of genetic material between bacteria### Phage Therapy

  • Phage therapy has several advantages, including:

    • Specificity: can target specific pathogens without disrupting normal bacteria flora
    • Nontoxic: phages are regularly consumed in foods and have been shown to be unintended contaminants in various medications and vaccines
    • Cheap: phages are the only medicine that multiplies, making them cost-effective
    • Rapid activity: phages have a high rate of success in treating bacterial infections
    • Resistance: while phage-resistant bacterial mutants can occur, most cases are less pathogenic
    • Evolve host range mutants: phages can evolve to target specific hosts, making them a unique medicine
  • However, phage therapy also has some drawbacks, including:

    • Narrow specificity: phages can be limited in their target range
    • Problems in certain industries (e.g. dairy industry)
    • Large number of unidentified ORFs (open reading frames)
    • Potential to carry virulence genes (although this is less likely with lytic phages)
    • Problems with early phage therapy, including exaggerated or unsubstantiated claims, poorly conceived experiments, and limited controls

Discovery of Bacteriophages

  • Bacteriophages were first discovered by E.H. Hankin in 1896, who reported that something in the waters of the Ganges River could pass through a fine porcelain filter and had marked antibacterial action
  • Frederick Tworth and Felix d'Herelle later recognized viruses that infect bacteria, which d'Herelle called "bacteriophages" (eaters of bacteria)

Phage Lifecycle

  • The phage lifecycle includes:
    • Infection: phages attach to and penetrate host cells
    • Destruction of bacterial DNA: phages break down the host cell's DNA
    • Replication of viral genome: phages replicate their own genetic material
    • Production of viral parts: phages assemble new viral particles
    • Packaging: phages package their genetic material into new viral particles
    • Lysis: phages lyse the host cell to release new viral particles

Variants of Phage Lifecycle

  • There are two types of phage lifecycles:
    • Lytic/virulent: phages always lyse infected cells
    • Temperate: phages lyse cells 50% of the time, and can be induced by certain chemicals, UV light, and antibiotics

Phage Behaving Badly

  • Phages can sometimes behave badly, including:
    • Lysogen-carrying bacteria: many bacteria carry phage genes, which can contribute to the virulence of the bacterium
    • Lethal for starter cultures: phages can infect and kill bacteria in starter cultures, such as those used in cheese and yogurt production

C.Difficile Infections

  • C.Difficile infections are a major problem, especially in hospitals, where:
    • Antibiotic use: inhibits the growth of beneficial bacteria, allowing C.Diff to overgrow
    • Hospital stays: increase the risk of colonization and transmission
    • C.Diff spores: can persist in the environment for many months and survive passage through the stomach
    • Early symptoms: include diarrhea, which can lead to aerosol and fecal contamination

Pathogenic Mechanism of C.Difficile

  • C.Difficile produces two main toxins: Toxin A and Toxin B, which:
    • Glucosylate (inactivate) a threonine residue on certain G proteins
    • Interfere with normal cellular function, leading to symptoms of pseudomembranous colitis

Treatment Strategies for C.Difficile

  • Current treatment strategies include:
    • Stopping antibiotics and administering vancomycin or metronidazole
    • Fecal transplants: reintroducing healthy flora to the gut
    • Phage therapy: targeting C.Difficile with specific phages

Prevention Strategies for C.Difficile

  • Prevention strategies include:
    • Reducing antibiotic use
    • Improving hospital hygiene and infection control
    • Using probiotics to promote healthy gut flora
    • Developing vaccines against C.Difficile

Test your knowledge on the transfer of plasmids between bacterial cells through conjugation. Explore how pili size can impact the efficiency of DNA transfer and how plasmids can acquire new resistances or virulence factors. Understand the process of replication and single-stranded transfer of conjugative plasmids.

Make Your Own Quizzes and Flashcards

Convert your notes into interactive study material.

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