L-14 Neoplasma II
41 Questions
0 Views

Choose a study mode

Play Quiz
Study Flashcards
Spaced Repetition
Chat to Lesson

Podcast

Play an AI-generated podcast conversation about this lesson

Questions and Answers

In DNA microarray technology, what is the primary purpose of labeling cDNA with fluorescent dyes?

  • To facilitate reverse transcription of mRNA into cDNA.
  • To enhance the specificity of cDNA binding to the microarray probes.
  • To enable detection and quantification of relative gene expression levels. (correct)
  • To prevent degradation of cDNA during hybridization.

What is the significance of performing reverse transcription (RT) in the context of DNA microarray analysis?

  • To convert unstable RNA into more stable DNA for analysis. (correct)
  • To directly amplify mRNA for hybridization.
  • To label mRNA with fluorescent dyes.
  • To degrade the original mRNA template.

During DNA microarray analysis, if a spot on the array shows a significantly stronger red fluorescence signal compared to green, what does this indicate about the corresponding gene?

  • The gene is expressed at a higher level in the reference sample compared to the test sample.
  • The gene is expressed at a higher level in the test sample compared to the reference sample. (correct)
  • The gene is silenced or not expressed in either sample.
  • The gene is equally expressed in both the test and reference samples.

What is the role of the DNA microarray in the overall process of gene expression analysis?

<p>To provide specific probes for hybridizing with the labeled cDNA. (D)</p> Signup and view all the answers

Why is it important to use two different fluorescent dyes (e.g., red and green) in DNA microarray experiments?

<p>To simultaneously compare gene expression levels between two different samples. (C)</p> Signup and view all the answers

Which of the following scenarios best describes the mechanism by which tumors exploit the CTLA-4 pathway to evade immune responses?

<p>Tumor cells induce dendritic cells to preferentially engage CTLA-4 over CD28, thereby inhibiting T-cell co-stimulation and antitumor response. (C)</p> Signup and view all the answers

In the context of cancer immunology, what is the primary role of immune checkpoints such as CTLA-4 and PD-1?

<p>To prevent excessive immune responses and autoimmunity by transmitting inhibitory signals to T-cells, maintaining immune homeostasis. (B)</p> Signup and view all the answers

A researcher is investigating a novel cancer immunotherapy that aims to enhance T-cell activation. Which of the following strategies would most likely achieve this goal by directly interfering with immune checkpoint pathways?

<p>Blocking the interaction between PD-1 and PD-L1 with a monoclonal antibody. (D)</p> Signup and view all the answers

How does the CTLA-4 pathway specifically impede effective antitumor immunity within the lymph node?

<p>By competing with CD28 for binding to B7 proteins on antigen-presenting cells, reducing the co-stimulatory signals required for T-cell activation. (C)</p> Signup and view all the answers

In the context of tumor immunology, what is the significance of the interaction between PD-L1 (Programmed Death-Ligand 1) expressed on tumor cells and PD-1 (Programmed Death-1) on T cells?

<p>It inhibits T cell activity, allowing tumor cells to evade immune detection and destruction. (B)</p> Signup and view all the answers

Which of the following describes the MOST significant difference between the mechanisms of action of CTLA-4 and PD-1 in suppressing antitumor immunity?

<p>CTLA-4 primarily affects T-cell activation in the lymph nodes, while PD-1 mainly affects T-cell activity in peripheral tissues and the tumor microenvironment. (A)</p> Signup and view all the answers

A pharmaceutical company is developing a new drug to enhance antitumor immunity. Clinical trials reveal that the drug effectively blocks the interaction between PD-L1 and PD-1, but some patients do not respond to the treatment. Which of the following mechanisms could BEST explain the lack of response in these patients?

<p>Upregulation of alternative immune checkpoint pathways, such as TIM-3 or LAG-3, which compensate for the PD-1/PD-L1 blockade. (D)</p> Signup and view all the answers

In sentinel lymph node biopsies, what is the primary purpose of using a probe?

<p>To follow radioactivity and detect the sentinel lymph node(s). (C)</p> Signup and view all the answers

What is the critical step after excising sentinel lymph nodes during a sentinel lymph node biopsy?

<p>Checking the nodes under a microscope for the presence of cancer cells. (C)</p> Signup and view all the answers

When are sentinel lymph node biopsies typically performed in relation to the removal of the primary tumor?

<p>Usually at the same time the primary tumor is removed. (B)</p> Signup and view all the answers

In Immunohistochemistry (IHC), what is the role of the enzyme-conjugated secondary antibody?

<p>To amplify the signal by binding to the primary antibody. (C)</p> Signup and view all the answers

What is the function of the chromogenic substrate, such as diaminobenzidine (DAB), in IHC?

<p>To catalyze the formation of a colored precipitate at the reaction site. (B)</p> Signup and view all the answers

What key information does IHC provide about a specific antigen (protein) in cells or tissues?

<p>The status (expression or not) and localization (membrane, cytoplasm, or nucleus). (B)</p> Signup and view all the answers

What is a primary application of IHC in the diagnosis of cancer?

<p>Identifying specific antigens (PROTEINS) within tissue sections. (D)</p> Signup and view all the answers

How does IHC contribute to identifying the site of origin for metastatic lesions?

<p>By detecting unique protein markers specific to certain tissues. (B)</p> Signup and view all the answers

In cancer diagnosis, what does "picking the needle out of the haystack" refer to in the context of IHC?

<p>Identifying rare cancer cells within a large sample of normal cells. (D)</p> Signup and view all the answers

How does IHC aid in assessing therapeutic significance in cancer?

<p>By detecting proteins that indicate whether a tumor will respond to a specific drug. (B)</p> Signup and view all the answers

In situ hybridization is primarily used to:

<p>Detect and localize specific nucleic acid sequences within cells or tissues. (C)</p> Signup and view all the answers

Which of the following best describes the function of probes in in situ hybridization?

<p>To bind selectively to specific DNA or RNA sequences of interest. (A)</p> Signup and view all the answers

What is the fundamental difference between radioactive in situ hybridization and chromogenic in situ hybridization?

<p>Radioactive methods use photographic emulsion and autoradiography for signal detection, while chromogenic methods use enzyme-substrate reactions to produce a colored precipitate. (B)</p> Signup and view all the answers

In chromogenic in situ hybridization, what is the role of horseradish peroxidase (HRP)?

<p>To catalyze a reaction that produces a visible colored precipitate. (D)</p> Signup and view all the answers

Why is autoradiography essential in radioactive in situ hybridization?

<p>To convert the radioactive signal into a visible image. (C)</p> Signup and view all the answers

What is the purpose of using diaminobenzidine (DAB) in chromogenic in situ hybridization?

<p>DAB serves as a substrate for an enzyme, resulting in a colored precipitate at the site of probe hybridization. (D)</p> Signup and view all the answers

In the context of cancer diagnosis, how are in situ hybridization techniques primarily utilized?

<p>To identify gene amplifications, deletions, or translocations. (D)</p> Signup and view all the answers

Which of the following scenarios would most likely warrant the use of in situ hybridization?

<p>Identifying the precise location of a specific mRNA transcript within a tissue sample. (C)</p> Signup and view all the answers

What is a key advantage of chromogenic in situ hybridization over radioactive in situ hybridization in modern diagnostic settings?

<p>Chromogenic methods are generally faster and do not require the handling of radioactive materials. (C)</p> Signup and view all the answers

Suppose a researcher is investigating a novel cancer biomarker using in situ hybridization. They observe a strong signal in the cytoplasm but not in the nucleus of cancer cells. What might this suggest about the biomarker?

<p>The biomarker is likely an mRNA transcript or a protein localized in the cytoplasm. (D)</p> Signup and view all the answers

What is the primary function of the CD3ζ (CD3 zeta) chain in T cell activation?

<p>Initiating and propagating the intracellular signaling cascade following antigen recognition. (C)</p> Signup and view all the answers

How do third-generation CARs enhance T-cell function compared to second-generation CARs?

<p>By incorporating additional co-stimulatory domains, potentially increasing persistence and potency. (C)</p> Signup and view all the answers

Which component of a CAR directly binds to a specific tumor antigen, such as CD19?

<p>The variable region of an antibody, often in the form of a single-chain variable fragment (scFv). (D)</p> Signup and view all the answers

What is the significance of engineering patient-derived CTLs with CARs outside the body before reinfusion?

<p>It provides a controlled environment to modify CTLs to target specific tumor antigens while minimizing harm to healthy cells. (C)</p> Signup and view all the answers

What is the role of the intracellular domain of the T cell receptor (TCR) in CAR T-cell therapy?

<p>To initiate and propagate the intracellular signaling cascade leading to T cell activation. (A)</p> Signup and view all the answers

Why might a 3rd generation CAR T-cell, with its additional costimulatory domains, be more effective than a 2nd generation CAR T-cell?

<p>The additional costimulatory domains can lead to increased T-cell persistence and potency, enhancing the anti-tumor response. (D)</p> Signup and view all the answers

What is the main reason leukapheresis is used in CAR T-cell therapy?

<p>To isolate and collect the patient’s T cells, which will be engineered to express CARs. (C)</p> Signup and view all the answers

A researcher is designing a CAR T-cell therapy for a specific B-cell lymphoma; the lymphoma cells express a unique variant of the CD19 protein not recognized by existing CAR T-cell therapies. What CAR component must be redesigned to ensure effective targeting of the lymphoma?

<p>The variable region of the antibody (scFv) to specifically bind the new CD19 variant. (C)</p> Signup and view all the answers

If a patient receiving CAR T-cell therapy experiences severe cytokine release syndrome (CRS) due to high levels of inflammatory cytokines, what aspect of the CAR T-cell design might be considered for modification in future therapies to mitigate this risk while maintaining efficacy?

<p>Incorporating a 'suicide gene' to allow for controlled depletion of CAR T-cells in case of severe toxicity. (C)</p> Signup and view all the answers

Flashcards

Tumor Heterogeneity

Tumors show variety in their cells and characteristics.

Immune Checkpoints

Proteins that regulate T-cell activation to prevent autoimmunity.

Examples of Immune Checkpoints

CTLA-4 and PD-1. When bound to ligands, they inhibit T-cell activation.

Ligands of CTLA-4 and PD-1

B7-1 (or B7-2) and PD-L1 (or PD-L2).

Signup and view all the flashcards

Tumor Immune Evasion via Checkpoints

Tumors use these pathways to avoid immune destruction.

Signup and view all the flashcards

CTLA-4 Mechanism

CTLA-4 prevents T-cell activation by binding B7, blocking CD28 co-stimulation.

Signup and view all the flashcards

T-cell Co-stimulation

Signal needed for full T-cell activation, often blocked by tumors.

Signup and view all the flashcards

Variable Region (scFv)

Part of an antibody that binds to a specific tumor antigen.

Signup and view all the flashcards

CD3ζ (CD3 zeta) Chain

The intracellular portion of the T cell receptor that triggers signaling, leading to T cell activation.

Signup and view all the flashcards

CD19

Found on the surface of B-cell leukemias and healthy B cells.

Signup and view all the flashcards

CAR T-Cell Therapy

CTLs (cytotoxic T lymphocytes) are modified to express CARs (chimeric antigen receptors) & reinfused into the patient.

Signup and view all the flashcards

CARs (Chimeric Antigen Receptors)

Engineered receptors that allow T cells to recognize and bind to specific antigens on tumor cells.

Signup and view all the flashcards

2nd Generation CAR

scFv, CD3ζ, and a single co-stimulatory domain (4-1BB or CD28).

Signup and view all the flashcards

3rd Generation CAR

Has an scFv, CD3ζ, and two co-stimulatory domains (e.g., CD27, CD28).

Signup and view all the flashcards

Co-stimulatory Domain

Enhances persistence and potency of CAR T-cells.

Signup and view all the flashcards

scFv (single chain variable fragment)

A fragment that contains the variable regions of both the heavy (VH) and light chains (VL) of an antibody, connected by a peptide linker.

Signup and view all the flashcards

DNA Microarrays

Also known as gene chips or DNA chips, used in molecular biology.

Signup and view all the flashcards

mRNA Extraction

Isolated from samples to analyze gene expression.

Signup and view all the flashcards

Reverse Transcription (RT)

Converts RNA to cDNA for amplification.

Signup and view all the flashcards

cDNA Labeling

Uses fluorescent dyes (red/green) to label cDNA from test and reference samples.

Signup and view all the flashcards

Hybridization in Microarrays

Labeled cDNAs bind to specific probes on the microarray to measure gene expression.

Signup and view all the flashcards

What is a probe?

A device used to detect radioactivity, often utilized in sentinel lymph node biopsies.

Signup and view all the flashcards

Sentinel Lymph Node

The first lymph node(s) to which cancer cells are likely to spread from a primary tumor.

Signup and view all the flashcards

Sentinel Lymph Node Biopsy

The removal and examination of sentinel lymph nodes to check for cancer cells.

Signup and view all the flashcards

Immunohistochemistry (IHC)

A technique used to detect specific antigens (proteins) in cells/tissues using antibodies.

Signup and view all the flashcards

Role of Enzymes in IHC

Enzymes like horseradish peroxidase (HRP) are used to oxidize chromogenic substrates, creating a colored precipitate at the reaction site, allowing visualization of the antigen-antibody complex.

Signup and view all the flashcards

Chromogenic Substrate

A chromogenic substrate produces a colored precipitate when oxidized by an enzyme, making the antigen-antibody reaction visible.

Signup and view all the flashcards

IHC Uses in Cancer

To diagnose tumors of unknown origin, identify the origin of metastases and detect proteins with prognostic or therapeutic significance.

Signup and view all the flashcards

What are Antigens in IHC?

Proteins that are detected by antibodies in IHC.

Signup and view all the flashcards

Role of the Primary Antibody

Antibodie's job is to bind to a specific antigen in a tissue sample.

Signup and view all the flashcards

Role of the Secondary Antibody

To bind to the primary antibody and amplify the signal through an enzymatic reaction.

Signup and view all the flashcards

In Situ Hybridization

A technique to find specific DNA or RNA sequences in cells within tissue.

Signup and view all the flashcards

In Situ Hybridization Probes

DNA segments that attach to specific sequences to identify gene changes.

Signup and view all the flashcards

In Situ Hybridization Use

Detects gene amplifications, deletions, or translocations in cancer cells.

Signup and view all the flashcards

Radioactive/Chromogenic ISH

Uses radioactive or chromogenic labels to visualize targeted sequences.

Signup and view all the flashcards

Fluorescence In Situ Hybridization (FISH)

Uses fluorescence to detect and visualize specific DNA sequences.

Signup and view all the flashcards

Radioactive ISH Process

Uses radiolabeled probes and photographic emulsion to detect signals.

Signup and view all the flashcards

Chromogenic ISH

Uses colored or visual probes, enzymes, and substrates to create a colored precipitate.

Signup and view all the flashcards

Chromogenic ISH Enzymes

Enzymes like HRP linked to streptavidin to detect biotin-labeled probes.

Signup and view all the flashcards

Chromogenic ISH Substrate

A substrate (DAB) reacts with an enzyme to create a colored precipitate.

Signup and view all the flashcards

Colored Precipitate in CISH

A colored deposit marking where the probe is bound to the DNA/RNA

Signup and view all the flashcards

Study Notes

Tumor Immunology

  • Paul Ehrlich theorized in 1909 that host defenses could prevent neoplastic cells from turning into tumors.
  • Lewis Thomas and Macfarlane Burnet coined the term immune surveillance in 1959-1970, suggesting the immune system constantly scans the body to destroy emerging cancer cells.
  • Lymphocyte infiltration in tumors often correlates with the outcome.
  • Tumors that were experimentally induced in animals are often immunogenic and can be eliminated by modulation of the immune system.
  • Immunodeficient people have a higher chance of developing some cancers.
  • New immunotherapies are showing impressive results in treating some cancers.

Immune Response against Tumors

  • Tumor-specific CTLs migrate back to and kill tumor cells.
  • Dendritic cells (DCs) phagocytose tumor cells or antigens and travel through afferent lymphatic vessels.
  • DCs present tumor antigens as class I MHC-associated peptides to naive CD8+ T cells in draining lymph nodes.

Tumor Antigens

  • Tumor antigens are presented on the tumor cell surface (or APC's cell surface) with Class I MHC to CD8+ cytotoxic T cells.
  • The immune system has inherent tolerance to normal proteins.
  • Tumor antigens also enter the class II MHC antigen-processing pathway in antigen presenting cells (APCs) and are recognized by CD4+ T cells.

Immune Evasion by Cancer

  • Cancer cells are destroyed by T cell recognition of tumor antigens leading to activation.
  • Escaping mechanisms involve clones outgrowth that lost tumor antigen or cannot present it with MHC class I molecules.

Immune Evasion by Cancer (Mechanisms Cont.)

  • Immune checkpoints are proteins which normally transmit inhibitory signals of T-cell activation to maintain immune responses within a normal physiological range and protect the host from autoimmune reactions.
  • Specific receptors like CTLA-4 and PD-1 bind to ligands B7-1 (or B7-2) and PD-L1 (or PD-L2), respectively which allows them to stop immune attacks.
  • CTLA-4 and PD-1/PD-L1 pathways are used by many tumors to evade immune attack.

CTLA-4 Pathway

  • Ineffective T cell response is caused by the upregulation of CTLA-4.
  • CTLA-4 competes with CD28 for B7 which impedes co-stimulation needed for CD8+ T cell activation.

CTLA-4 Immunotherapies

  • Blocking CTLA-4 with an antibody allows CD8+ T cells to be activated to kill tumor cells.
  • Ligands for CTLA-4 include B7-1 (aka CD80) and B7-2 (aka CD86).
  • Ipilimumab can now be used due to FDA approval in 2015, for metastatic cancers.

PD-1L Overexpression

  • Overexpression of PD-1 on T cells, or PD-1L or PD-L2 on tumor cells are Immune Evasion Mechanisms.

PD-1L Inhibition

  • Blocking the binding of PD-L1 or PD-L2 to PD-1 with antibodies allows T Cells to kill tumor cells.
  • Atezolizumab , FDA approved in 2016.
  • Pembrolizumab, FDA approved in 2014.

Chimeric Antigen Receptors (CARs)

  • Synthetic molecules that contain
    • An extracellular domain with variable regions of antibodies binding specific tumor antigens like CD19.
    • An intracellular domain with T cell receptor that recognize tumor antigens.
  • 2nd generation CAR: a single co-stimulatory domain, like 4-1BB, in addition to a CD3ζ signaling domain that makes the responses longer and more effective.
  • 3rd generation CAR: contains additional costimulatory domains like CD27 for increased persistency/potency.

CAR T-Cell Therapies

  • Engineered to express CARs by leukemia pheresis, then reinfused.
  • First CAR T-cell therapies target CD19 to treat ALL in kids, non-Hodgkin lymphoma, and diffuse large B-cell lymphoma.
  • Long-term remissions can be achieved, however "cytokine release syndrome" can occur due to CTL-released cytokines.

Pathologic and Laboratory Cancer Diagnosis

  • Morphologic Methods
  • Molecular Methods

Conventional Diagnosis of Solid Tumors

  • Consists of signs and symptoms like:
    • Palpable lump and/or pain
    • Fever, fatigue, weight gain or loss
    • Altered metabolism
  • Medical Imaging (X-rays, CT scan, MRI) can be performed
  • The Gold Standard is surgical biopsy followed by pathological diagnosis that consists of direct microscopic examination.

Sampling Approaches

  • Excisional biopsy (Bx) is removing the entire lesion.
  • Incisional Bx - a representative sample of the tumor.
  • Needle Bx is usually image-guided, with fine-needle aspiration.

Sampling Approaches (Cont.)

  • Cytology (Papanicolaou) smears show;
    • Large, flat cells with small nuclei (normal)
    • Sheet of cells with nuclear pleomorphism, hyperchromatic nuclei, frequent mitosis (M), and polymorphonuclear (PMN) cells. (abnormal)

Parameters for Neoplasm Diagnosis

  • Gross (macroscopic) tumor appearance.
  • Microscopic examination using H&E staining.
  • Immunohistochemistry for cell lineage identification or tumor type.
  • Cytogenetics/molecular approaches.

Macroscopic Examination

  • Considerations include adequate samples, labels on margins of incisions, open specimen sampling.
  • Pathologist's note consist of :
    • Consistency, color and extent
    • Shape of the neoplastic growth

Microscopic Examination

  • Uses Hematoxylin & Eosin [H&E] for classical tissue staining and Tissue Processing for Microscopic Examination, Paraffin-embedded.
  • Section with a microtome, mount on slide, then stain.

Microscopic examination with H&E

  • Frozen Sections (aka cryosections)
  • Mostly used for intraoperative diagnosis after rapid freezing.
  • Sections of ~ 1 µm are made in a cryostat (~microtome at -20 to -30°C).
  • Specimen is embedded in OCT compound, providing the proper matrix.
  • The process is rapid, but the quality is not as good as in paraffin-embedded specimens.

Frozen Section Uses

  • To verify identity of potential metastasis.
  • Mohs surgery- to remove thin skin layer to check for remaining cancer.

Frozen Sections are useful for

  • Procedure to test for cancer spread, most commonly to the lymph nodes (LN); Sentinel Lymph Node Biopsy (breast cancer, or melanoma).
    • Inject a radioactive substance, then use a probe, excise node, and check for cancer under a microscope.
    • Sentinel Lymph Node Biopsies are usually done when the primary tumor is removed.

Immunohistochemistry (IHC) in Tumor Diagnosis

  • Oxidization will cause the enzyme to form a colored precipitate at the reaction site
  • The IHC technique identifies,
    • Protein status expression
    • Localization in cell membranes/tissues
    • By use of specific antigens that visualize chromogen

IHC in Cancer

  • Identifies specific antigens (PROTEINS) within tissue sections.
  • Identifies malignant tumors of unknown histogenesis.
  • Identifies origin sites of metastatic lesions.
  • Detects proteins with prognostic or therapeutic significance.

IHC Cancer Markers

  • The tumor type depends on:
    • Carcinoma that contain cytokeratin
    • Sarcoma that contain vimentin
    • Melanoma that contain S-100 proteins
    • Lymphoma that contain CD45

More with IHC Cancer Markers, including:

  • Pan-CK positivity that can be used to trace undifferentiated tumors.
  • S-100 positivity can trace melanomas.

Tumor Marker Uses Include;

  • Support the diagnosis, with more tests
  • Reflects the stage/extent
  • Reflects the prognosis, through future evaluations

Tumor Markers detected on IHC

  • HER2/neu (erb-B2) is a more malignant Breast cancer, used for its therapeutic decision.
  • Estrogen and progesterone receptor (ER/PR) is for breast carcinoma used to predict the hormone therapy response.
  • Prostate-specific antigen (PSA) for metastatic prostate.
  • Carcinoembryonic antigen (CEA) is for Gl and lung adenoCa.
  • CA-19-9 for Gl and pancreatic CA.
  • CD117 (c-Kit) is for use in therapeutic decisions in GISTs.
  • Epidermal Growth Factor Receptor (EGFR) uses therapeutic decisions in Lung Ca.
  • HMB-45 melanoma detection and sentinel Nodes ID
  • Ki-67 measures Proliferative status

Immunophenotyping

  • Mostly used to classify leukemias and lymphomas:
    • T and B lymphocytes
    • Mononuclear-phagocytic cells
  • Fluorescent-labeled antibodies identify clusters of differentiation antigens (CDs) on cell surfaces.
  • DNA binding dyes analyze DNA content.

Cytogenetic Analysis

  • Primarily to confirm the diagnosis of certain hematological malignancies and solid tumors with chromosomal abnormalities.
    • CML(↑ Abl gene): t(9;22)-> PH chromosome
    • Burkitt lymphoma(↑ Myc gene): t(8;14)

Molecular Methods

  • In situ Hybridization is a technique used to detect and locate specific nucleic acid sequences in the nucleus (DNA) or cytoplasm (mRNA) on top of tissue section

In Situ Hybridization

  • Uses segments that are complementary to DNA/RNA to identify amplifications, deletions, or translocations.
  • Two main types of in situ hybridization:
    • Radioactive or Chromogenic
    • Fluorescence (FISH)

In Situ cont: a. Radioactive or Chromogenic

  • Radioactive probes (32P).
  • Photographic emulsion the signals.
  • Signal (black dots) autoradio images.
  • Chromogenic (biotin-labeled) probes.
  • Enzymes horseradish enhance signal.
  • Substrate precipitate visualization.

Molecular Diagnosis b. Fluorescence In Situ Hybridization (FISH)

  • Probes labeled with a fluorophore (FITC, fluorescein) allows detections with afluorescence microscope

In Situ Hybridization (FISH)

  • Multicolor FISH, 2 different fluorophores.
  • Abl (Chr 9)- will show red
  • Bcr can be stained by green

Molecular Diagnosis

  • Using Polymerase Chain Reaction, "PCR" which is; used to amplify a sequence with small sample sizes.
    • Involve specific primers to match nucleotide sequences
    • Forward and reverse primers (short single-strands of DNA) are designed to match the nucleotide sequence of short segments on each side of the target DNA.
    • Primers can serve as the starting points for DNA synthesis
    • Heat resistant polymerase (Taq) adds new DNA bases
    • PCR cycle involves 3 main process, those are:
      • Denaturing
      • Annealing
      • extension

PCR cont - Step 1 and 2

  • Denaturing is the double-stranded split into one strand at 95 C.
  • Annealing is temperatures cooling to attach primers on each end

PCR cont - Step 3 Extension

  • The end temperature is at 72 C add Taq polymerase, that makes new strands.
    • There are now billions of dsDNA pieces

DNA Microarrays

  • Unlike northern blotting, DNA microarrays allow a simultaneous expression changes in thousands of genes.Used to compare mRNA expression profiles (gene expression analysis) between different conditions, or mutations (genotyping) Tens of thousands DNA spots Each DNA or RNA is attached to a slide.

DNA Microarrays (cont.)

  • Reference samples for gene experiment are tested to extracted
  • RNA is complied by a transcription
  • Dyes determine amounts when scannned, red and green

Next Generation Sequencing

  • Enables identification of genetic mutations and is may aid in identifying with genomic DNA, for better diagnosis & assessment

Liquid Biopsies

  • Minimizes invasiveness of cancer biopsies
  • Can check for genetic and molecular characteristics of patients with liquid samples to determine cancer progression

Studying That Suits You

Use AI to generate personalized quizzes and flashcards to suit your learning preferences.

Quiz Team

Related Documents

Use Quizgecko on...
Browser
Browser