flashcard 16

(81 cards)

1
Q

What is analytical biochemistry?

A

Analytical biochemistry is the application of chemical techniques to separate, identify, and quantify the biochemical components in biological samples for purposes such as screening, diagnosis, prognosis, and monitoring.

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

What is clinical biochemistry?

A

Clinical biochemistry is an applied form of biochemistry focused on analysing bodily fluids (e.g., blood, urine, CSF) for diagnostic and therapeutic purposes, often organized into sub-specialties like routine chemistry, special chemistry, endocrinology, toxicology, therapeutic drug monitoring, urinalysis, and faecal analysis.

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

Name three sub-specialties of clinical biochemistry.

A

1) Clinical endocrinology (hormone analysis), 2) Toxicology (drugs of abuse and toxic chemicals), 3) Therapeutic Drug Monitoring (measuring medication levels to optimize dosage).

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

What tests are included in a basic metabolic panel?

A

Electrolytes (sodium, potassium, chloride, bicarbonate), blood urea nitrogen (BUN), creatinine, and glucose.

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

What additional tests are found in a comprehensive panel beyond the basic panel?

A

Calcium, liver function enzymes (ALP, ALT, AST), bilirubin, albumin, total protein, and any other tests tailored to specific clinical questions (e.g., thyroid, cardiac markers).

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

List the analytes measured in liver function tests (LFTs).

A

Total protein, albumin, globulins, A/G ratio, protein electrophoresis, urine protein, direct and indirect bilirubin (total bilirubin), AST, ALT, GGT, and ALP.

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

Why is protein electrophoresis performed as part of LFTs?

A

To separate serum proteins into fractions (albumin, α-, β-, γ-globulins), detect abnormal patterns (e.g., monoclonal gammopathy), and provide more detailed information than total protein alone.

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

Which enzymes are most sensitive indicators of hepatocellular injury?

A

ALT is most liver-specific; AST also rises in hepatocellular injury but can be elevated by muscle damage. GGT and ALP help distinguish hepatocellular injury from cholestasis.

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

Why is GGT measured alongside ALP?

A

Because GGT is specific to hepatobiliary injury, whereas ALP can also originate from bone or placenta; concurrent elevation of GGT and ALP confirms a hepatic source.

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

Name four common cardiac markers.

A

H-FABP, Troponin (I or T), Myoglobin, CK-MB, and B-type natriuretic peptide (BNP).

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

What is near patient testing (point-of-care testing)?

A

Testing performed at or near the site of patient care (e.g., bedside), offering faster results and requiring smaller samples but potentially less accuracy compared to central laboratory testing.

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

Give two examples of near patient tests.

A

Blood glucose meters and urine dipstick tests.

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

What are the advantages of near patient testing?

A

Faster turnaround time, smaller sample volumes, immediate clinical decision-making, and convenience for acute or ambulatory settings.

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

What are the disadvantages of near patient testing?

A

Potentially lower accuracy and precision, greater variability between operators, and limited test menus compared to centralized labs.

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

Which types of drugs commonly require therapeutic drug monitoring (TDM)?

A

Antibiotics, antiepileptics, cardiac drugs (e.g., digoxin), immunosuppressants (e.g., cyclosporine), antipsychotics, and anticoagulants (e.g., warfarin).

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

What factors affect drug availability in the body?

A

Absorption, distribution (protein binding, tissue sequestration), metabolism (phase I and phase II pathways), excretion (renal, biliary), dosing regimen, compliance, physiological differences (body mass, age), and drug interactions.

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

What is the difference between maximum tolerated dose (MTD) and minimum effective dose (MED)?

A

MTD is the highest dose that can be administered without unacceptable toxicity; MED is the lowest dose that produces the desired therapeutic effect. The therapeutic window lies between MED and MTD.

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

How many half-lives does it usually take to reach steady state drug concentration?

A

Approximately 4–5 half-lives.

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

What factors should be considered when designing a dosing regimen?

A

Peak and trough levels, half-life, therapeutic window, frequency of dosing, route of elimination, and patient-specific factors (renal/hepatic function, interactions).

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

What is quality control (QC) in clinical biochemistry?

A

Procedures to confirm validity of biochemical results by monitoring analytical performance, including internal QC (controls, reproducibility, drift) and external QC (proficiency testing, e.g., UKNEQAS).

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

What is a Levey‐Jennings chart used for?

A

To plot consecutive quality control measurements over time against mean and standard deviation limits, detecting trends (gradual shifts) or shifts (sudden deviations) in assay performance.

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

Define enzyme induction in drug interactions.

A

Enzyme induction occurs when a drug (inducing agent) increases the expression or activity of metabolic enzymes (e.g., CYP450), leading to faster metabolism of substrate drugs and reduced efficacy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Give an example of a clinically important enzyme induction.

A

Rifampicin induces CYP3A4, accelerating warfarin metabolism and reducing its anticoagulant effect, risking thrombosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Define enzyme inhibition in drug interactions.

A

Enzyme inhibition occurs when a drug inhibits the activity of metabolic enzymes, slowing metabolism of substrate drugs and increasing their plasma concentrations, potentially causing toxicity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Provide an example of enzyme inhibition used therapeutically.
Disulfiram inhibits aldehyde dehydrogenase, causing accumulation of acetaldehyde after alcohol intake, producing unpleasant effects that discourage alcohol consumption.
26
Why might inhibition of a prodrug-converting enzyme cause loss of drug activity?
Because the prodrug requires metabolic conversion (e.g., by CYP2C19) to its active form; inhibition prevents that conversion, rendering the drug ineffective (e.g., omeprazole inhibiting clopidogrel activation).
27
List four sources of individual variation affecting drug response.
Ethnicity (genetic polymorphisms), age (enzyme activity declines with age), pregnancy (increased cardiac output, GFR), and disease states (e.g., liver or kidney impairment).
28
What genetic variations can affect drug metabolism?
Polymorphisms in metabolic enzymes (e.g., CYP2D6 poor metabolizers) and plasma cholinesterase deficiency affecting succinylcholine metabolism or slow/fast acetylators affecting isoniazid clearance.
29
What is the principle of chromatography?
Separating components of a mixture based on their differential affinities for a stationary phase (solid or liquid) and a mobile phase (liquid or gas) passing over or through the stationary phase.
30
Name three major types of chromatography.
Gas chromatography (GC), liquid chromatography (LC/HPLC), and thin-layer chromatography (TLC).
31
What are two additional chromatographic methods used for proteins?
Ion-exchange chromatography and affinity chromatography (including hydrophobic interaction chromatography and chromatofocusing).
32
Give three forensic or food-related applications of small-molecule chromatography.
Detecting blood or accelerants in arson investigations (GC), monitoring pesticide residues in food (LC-MS), and verifying authenticity of products (TLC or HPLC fingerprinting).
33
What mobile and stationary phases are used in gas chromatography (GC)?
Mobile phase: inert carrier gas (e.g., helium); stationary phase: thin liquid film coated on the inside of a capillary column or packed in a column.
34
What is supercritical fluid chromatography (SFC)?
A separation technique using a supercritical fluid (e.g., CO₂ above its critical temperature and pressure) as the mobile phase, combining gas-like diffusivity with liquid-like solvation to separate small molecules.
35
Compare UV/Vis detection with diode array detection (DAD).
UV/Vis detection measures absorbance at a single selected wavelength; DAD captures absorbance across a range of wavelengths simultaneously, allowing spectral profiling but at higher cost.
36
Describe three types of mass spectrometry detectors mentioned.
Time-of-flight (TOF): separates ions by flight time in a field-free region; Triple quadrupole: uses three quadrupoles for precursor ion selection, collision-induced fragmentation, and product ion analysis; Orbitrap: traps ions in an electrostatic field, measures oscillation frequencies for high-resolution mass spectra.
37
What chromatographic variables affect column performance?
Column dimensions (length/width), particle size of packing (smaller particles increase resolution), adsorbent activity, column temperature (higher temperature accelerates elution but can affect selectivity), packing quality, and solvent quality (low viscosity improves efficiency).
38
Outline the general workflow of protein purification.
1) Extraction (lysis, homogenization, solubilization), 2) Fractionation (precipitation, centrifugation), 3) Purification strategies (chromatography: ion exchange, size exclusion, affinity, HIC, chromatofocusing), 4) Concentration (ultrafiltration, lyophilization), 5) Yield assessment and analysis (SDS-PAGE, Western blot).
39
How does ion-exchange chromatography separate proteins?
Proteins bind to oppositely charged groups on the resin based on their net charge at a given pH; elution is achieved by increasing ionic strength or changing pH to weaken protein-resin interactions (gradient elution).
40
What is size-exclusion chromatography and how does it separate proteins?
Also called gel filtration, it separates proteins by size: large molecules bypass resin pores and elute early (void volume), while smaller molecules enter pores and elute later, with retention volume related to molecular weight.
41
What key components define affinity chromatography?
A specific ligand immobilized on a matrix, often via a spacer arm to reduce steric hindrance; the target protein binds selectively to the ligand, then is eluted by changing buffer conditions or adding free ligand (step or gradient elution).
42
Describe hydrophobic interaction chromatography (HIC) and its elution strategy.
HIC uses a hydrophobic resin under high-salt conditions to strengthen hydrophobic interactions; decreasing salt concentration weakens these interactions and elutes proteins in order of increasing hydrophobicity.
43
What is chromatofocusing?
A form of ion-exchange chromatography where proteins are separated based on isoelectric point by creating an in-column pH gradient; proteins elute when the local pH equals their pI, enabling high-resolution separation.
44
What is the purpose of dialysis and desalting in protein purification?
Dialysis removes small contaminants (salts, free ligands) by diffusion through a semipermeable membrane; desalting columns or spin filters rapidly exchange buffer to remove unwanted small solutes.
45
How does SDS-PAGE separate proteins by molecular weight?
SDS denatures proteins and coats them with uniform negative charge (≈1.4 SDS molecules per amino acid), masking intrinsic charge and unfolding them into rodlike shapes so migration through the polyacrylamide gel depends primarily on molecular weight; smaller proteins migrate faster.
46
What information does isoelectric focusing (IEF) provide?
IEF separates proteins by isoelectric point: a pH gradient is established in a gel, and each protein migrates until it reaches the position where the gel pH equals its pI (net zero charge), allowing precise determination of pI.
47
What is two-dimensional electrophoresis (2D-PAGE)?
A technique combining IEF (first dimension, separation by pI) and SDS-PAGE (second dimension, separation by molecular weight) to resolve complex protein mixtures into individual spots for proteomic analysis.
48
How does Western blotting confirm protein identity?
Proteins separated by SDS-PAGE are transferred to a membrane, blocked, then probed with a primary antibody specific to the target protein followed by an enzyme- or fluorophore-conjugated secondary antibody; detection (chemiluminescence or fluorescence) reveals presence and relative abundance.
49
What are the advantages of ELISA for protein (antibody/antigen) detection?
High sensitivity and specificity (down to picogram levels), high throughput (96-well format), quantitative results, and ability to test various sample types (serum, plasma, urine, saliva).
50
What are the disadvantages of ELISA?
Temporary readouts due to enzyme/substrate kinetics requiring timely measurement, limited antigen information (only indicates presence or concentration, not structure), and potential cross-reactivity if antibodies are not highly specific.
51
Compare manual Sanger sequencing on slab gels versus automated capillary sequencing.
Manual Sanger: four separate PCR reactions with single ddNTPs, run on large slab polyacrylamide gels, slow, labor-intensive; Automated capillary: all ddNTPs mixed with unique fluorescent labels in one reaction, separated in narrow capillaries by automated machines with laser detection, faster, higher resolution, and automated readout.
52
What detection principle underlies absorption spectroscopy?
Absorption spectroscopy measures the decrease in light intensity at specific wavelengths as light passes through a sample; absorbance (A) is proportional to concentration (c) and path length (l) according to Beer–Lambert law (A = ε·c·l).
53
What factors can cause deviation from Beer–Lambert law in absorption measurements?
Light scattering (turbidity), reflection, refraction, matrix effects, high absorbance saturating the detector, and chemical interactions altering the absorptivity ε.
54
How is NADH used in enzyme kinetics measured by absorption spectroscopy?
NADH has a strong absorbance peak at 340 nm (ε ≈ 6,220 M⁻¹·cm⁻¹), whereas NAD⁺ does not. Monitoring the change in absorbance at 340 nm allows quantification of NADH formation or consumption over time.
55
How can absorption spectroscopy distinguish oxyhemoglobin from deoxyhemoglobin?
Oxyhemoglobin and deoxyhemoglobin have distinct absorbance spectra: the Soret band of oxyhemoglobin peaks near 415 nm with characteristic visible peaks, while deoxyhemoglobin’s peaks are shifted; comparing absorbance at specific λ max values differentiates them.
56
Define photoluminescence and its two main types.
Photoluminescence is light emission from a molecule after absorption of photons; it encompasses fluorescence (prompt emission with lifetimes of 10⁻¹⁰–10⁻⁸ s) and phosphorescence (delayed emission from a triplet state with lifetimes of 10⁻³–10² s).
57
What is quantum yield in fluorescence spectroscopy?
The quantum yield is the fraction of excited molecules that return to the ground state via fluorescence rather than nonradiative pathways.
58
What is quantum yield in fluorescence spectroscopy?
The quantum yield is the fraction of excited molecules that return to the ground state via fluorescence rather than nonradiative pathways, ranging from 0 (no fluorescence) to 1 (all excited molecules fluoresce).
59
How are excitation and emission spectra obtained in a fluorimeter?
Excitation spectrum: fix emission wavelength and scan excitation wavelengths, monitoring emitted intensity; Emission spectrum: fix excitation wavelength and scan emission wavelengths, recording intensity. Both spectra are corrected for lamp intensity and detector response.
60
What advantages does fluorescence spectroscopy offer over absorbance?
Higher sensitivity (detecting much lower analyte concentrations), greater selectivity (through distinct excitation/emission wavelengths), ability to probe molecular environments (e.g., polarity), and utility in imaging techniques like fluorescence microscopy.
61
What is a Stokes shift?
The Stokes shift is the difference between the peak excitation wavelength and the peak emission wavelength of a fluorophore, arising because excitation energy partially dissipates nonradiatively before emission, causing emission at longer wavelengths.
62
Define Förster Resonance Energy Transfer (FRET).
FRET is a distance-dependent energy transfer from an excited donor fluorophore to an acceptor molecule (fluorophore or quencher) within ~1–10 nm, requiring overlap between donor emission and acceptor absorption; FRET efficiency reports on molecular proximity.
63
What is fluorescence recovery after photobleaching (FRAP)?
FRAP measures molecular diffusion: a region of a fluorescently labeled sample is photobleached, then recovery of fluorescence (due to diffusion of unbleached fluorophores into the region) is monitored to quantify mobility or membrane fluidity.
64
What is bioluminescence and which enzyme is commonly used in reporter assays?
Bioluminescence is light emission from a biochemical reaction without external excitation; luciferase is commonly used to catalyze oxidation of luciferin (with ATP and O₂) to produce light, employed as a reporter for gene expression.
65
How is a luciferase reporter assay designed to measure transcriptional activation?
The promoter of interest is cloned upstream of the luciferase gene; when cellular factors activate the promoter, luciferase is expressed. Adding luciferin substrate produces light proportional to promoter activity, measured by a luminometer.
66
What is chemiluminescence and how is luminol used in forensic blood detection?
Chemiluminescence is light emission from a chemical reaction; luminol reacts with hydrogen peroxide (catalyzed by iron in hemoglobin) to form an excited intermediate that emits blue light upon returning to the ground state, revealing trace blood at crime scenes.
67
What are the three types of electrophoresis media and typical uses?
Agarose gels (0.5–2% agarose) for separating large DNA/RNA fragments in their native form; polyacrylamide gels (SDS-PAGE) for high-resolution separation of denatured proteins or small nucleic acids; capillary electrophoresis for high-resolution, rapid separation (e.g., DNA sequencing, small metabolites).
68
How does capillary electrophoresis differ from slab-gel electrophoresis?
Capillary electrophoresis uses narrow, buffer-filled capillaries and an electric field to separate analytes with high efficiency and speed, detecting fluorescently labeled fragments automatically; slab gels are larger, manual, and require staining or imaging.
69
What is energy level relaxation and how does it relate to fluorescence and phosphorescence?
After photon absorption (excitation to a higher energy state), molecules relax back to the ground state via internal conversion (nonradiative) or emission: fluorescence (singlet excited → singlet ground, fast) or phosphorescence (triplet excited → singlet ground, slow due to spin-forbidden transition).
70
What constitutes the excitation versus emission spectra of a fluorophore?
The excitation spectrum plots fluorescence intensity while varying excitation wavelength at a fixed emission wavelength (similar to absorbance). The emission spectrum plots intensity of emitted light while varying emission wavelength at a fixed excitation wavelength.
71
Why must samples be clear and non-turbid for fluorescence measurements?
Turbidity causes light scattering, which diverts excitation and emission photons out of the detection path, reducing measured fluorescence intensity and distorting spectral data.
72
What is circular dichroism spectroscopy used for in protein analysis?
Circular dichroism measures differential absorption of left- versus right-circularly polarized light by chiral molecules, providing information on protein secondary structure (α-helix, β-sheet content).
73
How does confocal fluorescence microscopy achieve enhanced resolution?
By using point illumination and a pinhole to reject out-of-focus fluorescence, confocal microscopy collects sharp optical sections at various depths, enabling 3D reconstruction with high contrast and resolution.
74
What wavelength range defines near-infrared (NIR) absorption spectroscopy?
Approximately 800–2,500 nm (12,500–4,000 cm⁻¹), capturing overtone and combination vibrational bands useful for compositional analysis of liquids, solids, and biological samples.
75
Why does water appear blue according to NIR absorption spectroscopy?
Water strongly absorbs red light (>600 nm); because longer red wavelengths are absorbed, only shorter (blue) wavelengths transmit through sufficiently in large bodies, making water appear blue.
76
How can heavy water (D₂O) be distinguished from normal water by NIR?
D₂O has vibrational absorption bands shifted to longer wavelengths (lower wavenumbers) compared to H₂O; measuring absorption around 2,000 nm can differentiate heavy water from normal water.
77
What is a major limitation of X-ray crystallography?
It requires well-ordered protein crystals, and many proteins are difficult or impossible to crystallize, precluding structural determination by X-ray methods.
78
What information does NMR spectroscopy provide for proteins?
NMR yields chemical shift and coupling data for NMR-active nuclei (¹H, ¹³C, ¹⁵N, ¹⁹F, ³¹P), allowing determination of atomic-level structures in solution, measurement of interatomic distances, and monitoring dynamics (folding, ligand interactions).
79
Why is NMR advantageous over crystallography for studying protein dynamics?
NMR analyses proteins in solution, enabling observation of conformational changes, folding/unfolding, and protein–ligand interactions in a near-physiological environment, whereas X-ray crystallography only reveals static, crystalline structures.
80
How does clinical biochemistry apply spectroscopy in diagnostics?
Absorbance assays measure enzyme activities and metabolite concentrations (e.g., NADH at 340 nm, bilirubin, creatinine); fluorescence assays quantify low-abundance biomarkers; chemiluminescent immunoassays detect hormones and proteins with high sensitivity.
81
Summarize the key steps to prepare for an analytical and clinical biochemistry exam.
Review lecture slides and practical notes for each technique (TDM, chromatography, electrophoresis, spectroscopy), understand principles and clinical applications, memorize analyte panels (LFTs, electrolytes, cardiac markers), and practice interpreting QC charts, drug interaction scenarios, and example questions.