ANTISERA Flashcards

1
Q

Who laid laid the foundation for immunology?

A

Emil van Behring had
shown that immunization
created a blood-born
factor that could be
transferred to protect
against diphtheria. by demonstrating that immunization could produce a blood-borne factor, later identified as antibodies, which could be transferred to provide protection against diphtheria.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

what is immunization

A

Immunization is the process by which an individual’s immune system becomes fortified against an agent (known as the immunogen). When the immune system is exposed to molecules that are foreign to the body, called antigens, it triggers an immune response. The process of immunization can occur naturally or artificially

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is an Immunoassay?

A

An immunoassay is a biochemical test that measures the presence or concentration of a substance, often an antigen, in a solution through the use of an antibody or antibodies. Immunoassays for SARS-CoV-2 antigens are designed to detect specific proteins from the virus, typically the nucleocapsid or spike proteins, in samples such as nasal or throat swabs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Lateral Flow Assays (LFAs)

A

Also known as rapid antigen tests, these are quick and easy-to-use tests that can be performed at the point of care. They typically provide results within 15-30 minutes.
The test strip contains antibodies that bind to the SARS-CoV-2 antigens if they are present in the sample. A visible line appears on the strip to indicate a positive result.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Enzyme-Linked Immunosorbent Assays (ELISAs)

A

These assays are more sensitive and are usually performed in a laboratory setting. They involve multiple steps where the antigen binds to an antibody attached to a solid surface, and then a secondary antibody linked to an enzyme reacts to produce a detectable signal.
ELISAs can quantify the amount of viral antigen present, providing more detailed information compared to rapid tests

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is a GMO Immunoassay?

A

An immunoassay for GMO detection is a biochemical test that uses antibodies to identify and measure specific proteins that are unique to genetically modified organisms. These proteins are typically those expressed by the inserted genetic material in the GMO.

immunoassay test for the CP4 protein that makes crops tolerant to Roundup® herbicides.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are Food Allergen Tests?

A

Food allergen tests are designed to detect specific proteins in food that can cause allergic reactions in sensitive individuals. These tests use antibodies that bind to these allergenic proteins, allowing for their identification and quantification.

Types of Antibody-Based Food Allergen Tests
Enzyme-Linked Immunosorbent Assays (ELISAs):

ELISAs are widely used for detecting food allergens. They involve binding allergen-specific antibodies to a solid surface, applying the food sample, and then adding a secondary antibody that produces a measurable signal, typically a color change.
ELISAs can be both qualitative (indicating the presence or absence of an allergen) and quantitative (measuring the amount of allergen).
Lateral Flow Devices (LFDs):

Also known as rapid tests or strip tests, these are user-friendly and provide quick results. A sample is applied to a test strip, and if the allergen is present, it binds to antibodies on the strip and produces a visible line.
LFDs are typically used for on-site testing and are qualitative or semi-quantitative.
Western Blotting:

This technique involves separating proteins by gel electrophoresis, transferring them to a membrane, and detecting specific allergens using antibodies.
Western blotting is highly specific and is used for confirming the presence of allergens detected by other methods.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

BioKits

A

BioKits are a series of antibody-based test kits specifically designed for detecting various food allergens and genetically modified organisms (GMOs) in food products. These kits leverage immunoassay technology to provide reliable, rapid, and user-friendly testing solutions for the food industry.

The kits are designed to detect specific proteins (allergens) or genetic modifications (GMOs) in food samples, ensuring compliance with labeling regulations and safeguarding consumer health.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Antibody-Based Applications for Raw Meat Analysis

A

Enzyme-Linked Immunosorbent Assays (ELISAs): ELISAs can detect pathogens like Salmonella, Escherichia coli (E. coli), Listeria monocytogenes, and Campylobacter.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Where are antibodies used in everyday life?

A

conventional pregnancy tests
* rapid tests in the doctor’s office
* at home

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

what are some Antibody-based applications?

A

-In vivo therapy
-In vivo diagnostic
-Vaccination
-Chromatography
(t.ex. affinity chromatography)
-Biosensors
(e.g. continous-flow measuremets in biosensors)
-Immunoblotting techniques
-Immunoprecipitation
(e.g. studies of protein-protein interactions)

-Immunofluorescence/histology
-Protein-DNA interactions
-Microarray techniques
(focussed assay to proteome-scale
analysis)
-Flow cytometry
-Qualitative analysis
(pregnancy test, drug test, HIV test,
bacterial infections)
-Quantitative analysis
(precipitation/nefelometri, competitive
analysis, non-competitive analysis)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

what is In vivo therapy

A

In vivo therapy refers to medical treatments and interventions performed directly within a living organism. This term encompasses various therapeutic approaches where the treatment is administered in the body, as opposed to in vitro therapies, which are conducted outside the body in a controlled laboratory environment. In vivo therapies can target diseases at the molecular, cellular, or systemic level and include a broad range of techniques and applications.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

In vivo diagnostic

A

In vivo diagnostics refer to medical diagnostic techniques and procedures performed directly within a living organism to detect and monitor diseases or medical conditions. These methods enable real-time assessment of biological processes in their natural context, providing valuable information for accurate diagnosis, disease progression monitoring, and treatment efficacy evaluation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Vaccination

A

Vaccination works by introducing antigens (substances that the immune system recognizes as foreign) into the body. These antigens are typically derived from the pathogen itself, either in a weakened or inactivated form, or as parts of the pathogen such as proteins or sugars. The immune system responds to these antigens by producing antibodies and memory cells. If the vaccinated individual is later exposed to the actual pathogen, their immune system can quickly recognize and attack it, preventing illness.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Chromatography
(t.ex. affinity chromatography)

A

It involves the separation of components in a mixture based on their differential distribution between two phases: a stationary phase and a mobile phase. One particularly useful type of chromatography is affinity chromatography, which selectively purifies target molecules based on their specific interactions with immobilized ligands on the stationary phase.

Antibody Purification: Antibodies can be purified from serum or cell culture supernatants using affinity chromatography columns containing immobilized protein A or protein G

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Biosensors
(e.g. continous-flow measuremets in biosensors)

A

Antibody-based biosensors are analytical devices that utilize antibodies as recognition elements to detect specific target molecules, such as proteins, pathogens, or environmental contaminants. These biosensors convert the binding events between antibodies and target molecules into measurable signals, providing rapid and sensitive detection capabilities. Continuous-flow measurements in biosensors are a common approach to enable real-time monitoring of target analytes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Immunoblotting techniques

A

also known as Western blotting, is a widely used laboratory technique for detecting specific proteins in a complex mixture of proteins extracted from cells or tissues. This technique combines gel electrophoresis for protein separation with antibody-based detection for protein identification.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Immunoprecipitation
(e.g. studies of protein-protein interactions)

A

isolation and purification of specific proteins or protein complexes from complex biological samples, such as cell lysates or tissue homogenates, based on their interactions with specific antibodies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Immunofluorescence/histology

A

Immunofluorescence (IF)
Principle: Immunofluorescence utilizes the selective binding of fluorescently labeled antibodies to specific target molecules (e.g., proteins) within cells or tissues.

Procedure:

Sample Preparation: Cells or tissue sections are fixed, permeabilized (if necessary), and blocked to preserve cellular structures and reduce nonspecific binding.
Primary Antibody Incubation: Samples are incubated with primary antibodies specific to the target molecule(s) of interest.
Secondary Antibody Incubation: Fluorescently labeled secondary antibodies, which recognize the primary antibodies, are applied to the samples.
Washing: Unbound antibodies are removed by washing steps to reduce background fluorescence.
Mounting: Samples are mounted with a mounting medium containing fluorescent dyes (e.g., DAPI for nuclear staining) to visualize cell structures.
Microscopic Imaging: The samples are examined using a fluorescence microscope to visualize the fluorescently labeled target molecules within cells or tissues.
Applications:

Visualization and localization of proteins, nucleic acids, and other molecules within cells and tissues.
Studies of cellular morphology, subcellular structures, and protein-protein interactions.
Diagnostic applications in pathology, immunology, and microbiology.

Histology
Principle: Histology involves the preparation of tissue samples for microscopic examination to study the structure, composition, and organization of tissues and organs.

Procedure:

Tissue Fixation: Tissues are fixed using chemical fixatives (e.g., formaldehyde) to preserve cellular structures and prevent decay.
Tissue Processing: Fixed tissues are dehydrated, embedded in a solid medium (e.g., paraffin wax or resin), and sectioned into thin slices (histological sections).
Staining: Sections are stained using various dyes or stains to highlight different tissue components, such as nuclei (hematoxylin), cytoplasm (eosin), connective tissue (Masson’s trichrome), and specific cell types (immunohistochemical staining).
Mounting: Stained sections are mounted onto glass slides and coverslipped for microscopic examination.
Microscopic Imaging: The prepared slides are examined using a light microscope to visualize tissue structures and patterns.
Applications:

Diagnosis of diseases and pathological conditions (histopathology).
Research studies of tissue development, morphology, and pathology.
Forensic analysis and comparative anatomy studies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

protein-DNA interactions

A

Techniques such as ChIP, EMSA, immunofluorescence, and immunohistochemistry provide insights into the dynamics, localization, and functional consequences of protein-DNA interactions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Microarray techniques
(focussed assay to proteome-scale
analysis)

A

Tissue Microarrays (TMAs):

Principle: Tissue microarrays are constructed by embedding multiple tissue samples into a single paraffin block. Thin sections are cut from the block, allowing simultaneous analysis of multiple tissue specimens on a single slide.
Procedure: Tissue sections are immunostained with specific antibodies, and protein expression patterns are analyzed by microscopy.
Applications: Studying protein expression in clinical samples, biomarker validation, cancer research.
Reverse Phase Protein Arrays (RPPAs):

Principle: RPPAs are used to analyze protein expression and post-translational modifications in large sample cohorts.
Procedure: Protein lysates from samples are printed onto a solid support as a microarray. The microarray is probed with specific antibodies, and protein levels are quantified using fluorescence or chemiluminescence.
Applications: Biomarker discovery, drug development, cancer research.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Flow cytometry

A

to analyze and quantify various properties of individual cells within a heterogeneous population. This technique utilizes fluorescently labeled antibodies to detect specific cell surface markers, intracellular proteins, and other molecules of interest

23
Q

Qualitative analysis
(pregnancy test, drug test, HIV test,
bacterial infections)

A

Antibody-based qualitative analysis refers to tests that provide a yes/no answer, indicating the presence or absence of specific analytes (such as hormones, drugs, or pathogens) in a sample. These tests leverage the high specificity of antibodies to detect target molecules and are widely used in various fields, including medical diagnostics, environmental testing, and food safety

24
Q
A

Principle:

These techniques measure the amount of immune complex formed between antibodies and antigens in a solution. The complex can be measured by its ability to scatter light (nephelometry) or form a precipitate (precipitation).
Precipitation:

Procedure: Antigen and antibody solutions are mixed. At optimal ratios, they form an insoluble complex that precipitates out of solution.
Application: Measuring immunoglobulins, complement components, and other serum proteins.
Nephelometry:

Procedure: The sample is mixed with specific antibodies, and the immune complex formation causes light scattering. The amount of scattered light is proportional to the concentration of the antigen.
Application: Quantifying proteins such as C-reactive protein (CRP), immunoglobulins, and complement components.
Competitive Analysis
Principle:

In competitive assays, an unknown amount of antigen in the sample competes with a known quantity of labeled antigen for binding to a limited number of antibody binding sites.
Procedure:

A fixed amount of labeled antigen and the sample containing the unknown antigen are incubated with a specific antibody.
The antibody binds to both labeled and unlabeled antigens. The more unlabeled antigen present in the sample, the less labeled antigen will bind to the antibody.
The amount of labeled antigen-antibody complex is inversely proportional to the concentration of the antigen in the sample.
Detection is typically done using a signal from the labeled antigen, such as radioactivity, fluorescence, or enzyme activity.
Applications:

Measuring small molecules like hormones (e.g., cortisol, thyroxine), drugs (e.g., therapeutic drug monitoring), and toxins.
Non-Competitive Analysis (Sandwich ELISA)
Principle:

Non-competitive assays, often referred to as sandwich assays, measure the antigen concentration by capturing the antigen between two antibodies: a capture antibody and a detection antibody.
Procedure:

Capture Phase: The sample is added to a plate coated with a capture antibody specific to the antigen of interest.
Binding Phase: The antigen in the sample binds to the capture antibody.
Detection Phase: A second antibody, specific to a different epitope on the antigen and conjugated with a detectable label (enzyme, fluorophore, etc.), is added. This forms a “sandwich” with the antigen in between the two antibodies.
Signal Measurement: The bound detection antibody’s label produces a measurable signal proportional to the antigen concentration.
Applications:

Measuring large proteins, cytokines, growth factors, and other biomolecules in clinical samples (e.g., blood, urine, saliva).

25
Q

Examples of relevant SARS-CoV-2 assays

A
  • PCR etc.
    -detect viral nucleic acid (ongoing infection)
  • Immunoassay
  • detect virus-specific IgM (recent infection)
  • detect increase in virus-specific IgG (recent/ongoing infection)
  • detect virus-specific IgG/IgA (past infection/protection)
  • detect the virus (antigen test)
    *-study the biology of the virus
26
Q

what are Sources of antibodies – development path?

A

Immunization resulting in polyclonal antisera or
purified fractions there of

Monoclonal antibody technology

Recombinant antibody technology

Combinatorial library technology
such as phage display, often in combination with
recombinant antibody technology

27
Q

what is the goal of antisera/using antiserum?

A

To stimulate the immune system to
produce antibodies with:
i) High affinity (binding strength)
ii) High specificity

28
Q

key players in antisera?

A

IN = IMMUNOGEN ->
B CELLS <->
T-cells <- APC
OUT= ANTIBODIES

Process - Immunization
Def: deliberate provocation of an adaptive immune response.

29
Q

What is the difference between an immunogen and an antigen?

A

Immunogen – any molecule that can elicite an adaptive immune response on injection into a person or animal.

Antigen – any molecule that can specifically bind to an antibody (or a T cell receptor).

NB! An antigen does not have to be an immunogen.

30
Q

describe the Process how antisera is made

A

Antigen preparation incl. coupling of B / T cells epitop (if needed)
Immunization
–>
Uptake of antigen by APC
–>
Presentation of antigen for T cells
(MHC class II/peptid - T cell receptor)
–>
Interaction between specific B and T cells
–>
Stimulation of B cells to antibody production
and affinity maturation
–>
Antisera composed of good antibodies!

31
Q

Antibody-based reagents should be generated
while considering the area of application, what different properties can the antibody have?

A
  • Polyclonal / monoclonal antibodies
  • Fine specificity
  • Cross-reactivitites (the binding of an antibody to an antigen not used to
    elicit that antibody)
32
Q

Polyclonal ab

A

several ab against different epitopes

33
Q

Monoclonal ab

A

one ab against one epitope

34
Q

STRATEGY FOR PRODUCTION OF ANTISERUM

A

1) Preparation of immunogen
- Purification / control of purif.
- Coupling to carrier (if needed).
2) Immunization
- Choice of animal
- Schema (e.g. Booster)
- Adjuvant
- Amount (Dose)
- Boost
3) Antiserum retrieval
4) Testing & modification
- Specificity
- Titer
- Binding properties
- Absorption
5) Purification
- ammoniumsulphate precipitation
- ion-exchange chromatography
- protein A / protein G
- affinity chromatography
- etc
6) Quality control
- potential application
- isotype
- stability
- specificity
- affinity
- etc

35
Q

Hapten

A

Low-molecular weight antigen
(that elicit an immune response
only after it has been coupled
to a carrier)

36
Q

Immunogen – important factors?

A

Purity
Correct structure (protein folding)
Linear / discontinous epitopes
Which epitopes are exposed?

37
Q

Increased immunity with increased size?

A

*High-molecular weight (protein) antigen
Results in adequate immunization

  • Hapten – low-molecular weight antigen
    Mainly results in NO immunization
    (common feature - no T cell activation)
38
Q

How do you make low-molecular weight substances and non-immunogenic substaces immunogenic ?

A

Couple B- and T-cell epitopes !
i.e. many low-molecular weight substances do not
elicit an immune response (antibodies) since they do
not react with both B and T cells.

Ab response!
Hapten - carrier

39
Q

how to make the hapten immunogenic?

A

Choice of carrier molecule:

Linking the hapten to T cell epitope containing structure makes the
hapten immunogenic.
1. Protein (albumin etc.)
2. MDP (muramyl dipeptid)
3. Synthetic peptid.
etc

40
Q

Hapten

A

(from the Greek haptein – to fasten)

41
Q

Choice of coupling point for hapten?

A

Choice of coupling point will influence the specificity of the antisera.
Structures far from the coupling point are best recognized by the induced
antibodies.
Different coupling chemistries can be used, e.g. carbodiimid (t.ex. EDC)
N-hydroxyl succinimid ester (NHS)

42
Q

Issue – selective analysis of several very similar peptides possible ?
i.e. impact of coupling strategy on ab specificity

A

Analyte (used for immunization)
Naturally occuring hormone
Another interesting analyte
Potential degradation products

Selective analysis of several very similar peptides, especially those that differ by only a few amino acids, poses a significant challenge in immunoassay development. The coupling strategy used during antibody production and the specificity of the antibodies are crucial factors that impact the ability to differentiate between these peptides

43
Q

Factors Influencing Selective Analysis
Antibody Specificity:

A

Epitope Selection: The choice of epitope, or the specific part of the peptide that the antibody recognizes, is critical. Highly specific epitopes that capture unique features of each peptide are necessary for selective detection.
Antibody Affinity: High-affinity antibodies are generally more specific, but if the affinity for similar peptides is too high, cross-reactivity can occur.
Coupling Strategy:

Carrier Proteins: When peptides are conjugated to carrier proteins (e.g., KLH, BSA) for immunization, the site of conjugation can affect the antibody’s specificity. Conjugation should avoid masking the unique epitopes.
Chemical Modifications: Using different chemical linkers or spacer arms can influence the accessibility and presentation of epitopes, impacting antibody specificit

Antibody Production:

Monoclonal vs. Polyclonal Antibodies: Monoclonal antibodies are more specific than polyclonal antibodies because they recognize a single epitope. Polyclonal antibodies may recognize multiple epitopes, increasing the risk of cross-reactivity.
Phage Display and Hybridoma Technology: Techniques like phage display can be used to screen for antibodies with high specificity to distinct peptides.

44
Q

The amount (dose) of antigen is essential.

A

Not to low, not to high – moderate (”lagom”) is best!

45
Q

How is the immunogen administrated?

A

<–Increased immunogenicity

Subcutaneous (sc)> intraperitoneal (ip)> intravenous or intragastic
(iv)

46
Q

IP injection

A

is the injection of a substance into the peritoneum (body cavity).

47
Q

Repeated immunization results in maturation
and enhanced levels of antibodies, what happens?

A

IgM –> IgG
Low affinity –> high affinity
Low conc. –>high conc.

48
Q

Adjuvant

A

compound(s) that enhances the immune response for an antigen
when mixed and administrated together.

49
Q

Effects of Adjuvants?

A

Increases the ab response due to more effective ag presentation; reduces
the required dose of antigen.
*Increases the immunogenicity
*May alter the isotype profile in the ab response
*Storage-effect – gives a prolonged ab response
Note:
Increase the immunogenicity by
i) Converts soluble proteins to particulate material, that is more easily taken up by APCs.
ii) Presence of bacteria/bacterial material stimulates immune system.

50
Q

Choice of animal host for antibody production

A

Different response towards certain antigen in different animals
Demands on the amount of antibody to be generated
Demands on the effector functions (e.g. ab isotype)
Commonly used animal hosts
Mouse, rabbit, goat, chicken

51
Q

Specificity issues?

A

Does your antibody really bind specifically?
* Does it bind what it is supposed to bind?
* Does it bind things that it is not supposed to bind?
* Is your antibody specific in all applications if proven
specific in one application?

52
Q

what is Cross-reactivity

A

The ability of the antibody to react with antigen(s) other than the one used to elicit the original antibody response.

Identical epitopes.
”Sufficiently similar” epitopes. (different binding affinities?)

53
Q

Absorption

A

a process in which the
population of cross-reactive antibodies are
removed (e.g. by affinity chromatography)