All Topics
biology-9700 | as-a-level
Responsive Image
Uncontrolled cell division and tumours

Topic 2/3

left-arrow
left-arrow
archive-add download share

Your Flashcards are Ready!

16 Flashcards in this deck.

or
NavTopLeftBtn
NavTopRightBtn
3
Still Learning
I know
12

Uncontrolled Cell Division and Tumours

Introduction

Uncontrolled cell division is a fundamental process underlying the formation of tumours, a critical topic in the study of the mitotic cell cycle. Understanding this phenomenon is essential for AS & A Level Biology (9700) students, as it provides insights into the mechanisms of cancer development, diagnosis, and treatment. This article delves into the intricacies of how normal cellular processes can go awry, leading to malignancies, and explores the biological principles that govern these changes.

Key Concepts

Cell Cycle Regulation

The cell cycle is a series of phases that a cell undergoes to grow and divide. It consists of four main stages: G1 (gap 1), S (synthesis), G2 (gap 2), and M (mitosis). Regulation of the cell cycle ensures that cells divide only when necessary and that each phase is accurately completed before proceeding to the next. This regulation is orchestrated by a network of proteins, including cyclins and cyclin-dependent kinases (CDKs), which act as checkpoints to monitor the cell's status.

Cyclins are proteins whose levels fluctuate throughout the cell cycle, activating CDKs at specific stages. For example, Cyclin D binds to CDK4/6 during G1, facilitating the transition to the S phase. Similarly, Cyclin B-CDK1 complexes drive the cell into mitosis. These checkpoints are critical for preventing the replication of damaged DNA and ensuring genomic integrity.

Mechanisms of Uncontrolled Cell Division

Uncontrolled cell division occurs when regulatory mechanisms fail, allowing cells to proliferate unchecked. This can result from mutations in genes that control the cell cycle, such as oncogenes and tumor suppressor genes. Oncogenes are mutated forms of normal genes (proto-oncogenes) that promote cell division. When activated, they can lead to excessive proliferation. Examples include the RAS and MYC oncogenes, which are implicated in various cancers.

Tumor suppressor genes act as brakes on cell division. When these genes are inactivated or lost due to mutations, cells can divide uncontrollably. The TP53 gene, encoding the p53 protein, is a well-known tumor suppressor. p53 plays a crucial role in DNA repair, inducing cell cycle arrest or apoptosis in response to DNA damage. Loss of p53 function compromises the cell's ability to prevent the propagation of genetic abnormalities.

DNA Replication and Mutation Accumulation

Accurate DNA replication is vital for maintaining genetic stability. However, errors during replication can lead to mutations, which may accumulate over time. Mutations in key regulatory genes can disrupt normal cell cycle control, promoting uncontrolled division. For instance, mutations that confer resistance to apoptosis allow cells with damaged DNA to survive and proliferate, contributing to tumour formation.

Exposure to carcinogens, such as tobacco smoke or UV radiation, can increase the mutation rate by causing DNA damage. Reactive oxygen species (ROS) generated during cellular metabolism can also damage DNA, proteins, and lipids, further exacerbating genetic instability. The accumulation of such mutations is a hallmark of cancer and is central to the development of malignant tumours.

Signal Transduction Pathways

Cell division is regulated by complex signal transduction pathways that respond to internal and external cues. These pathways involve the transmission of signals from cell surface receptors to the nucleus, resulting in changes in gene expression. The mitogen-activated protein kinase (MAPK) pathway is one such pathway that regulates cell growth, differentiation, and division.

Dysregulation of signal transduction pathways can lead to persistent activation of growth-promoting signals, contributing to uncontrolled cell division. For example, mutations that lead to constitutive activation of the MAPK pathway can drive continuous cell proliferation, bypassing normal regulatory checkpoints and fostering tumour growth.

Apoptosis and Its Inhibition

Apoptosis, or programmed cell death, is a mechanism by which damaged or unnecessary cells are eliminated. It serves as a crucial counterbalance to cell division, maintaining tissue homeostasis. Key proteins involved in apoptosis include the Bcl-2 family and caspases, which orchestrate the dismantling of cellular components.

In cancerous cells, the apoptotic pathways are often disrupted, allowing cells with genomic abnormalities to survive and proliferate. Overexpression of anti-apoptotic proteins, such as Bcl-2, or loss of pro-apoptotic factors, impairs the cell's ability to undergo apoptosis, facilitating the accumulation of malignant cells.

Angiogenesis and Tumour Growth

Angiogenesis, the formation of new blood vessels, is essential for tumour growth beyond a certain size. Tumours require a blood supply to obtain nutrients and oxygen and to remove waste products. Vascular endothelial growth factor (VEGF) is a key mediator of angiogenesis, promoting the proliferation and migration of endothelial cells to form new blood vessels.

Uncontrolled cell division within tumours stimulates the production of VEGF, enhancing angiogenesis and enabling the tumour to expand. Additionally, angiogenesis facilitates metastasis by providing a route for cancer cells to enter the bloodstream and colonize distant organs, exacerbating the malignancy.

Genetic Instability

Genetic instability refers to an increased rate of mutations within the genome, contributing to cancer progression. It arises from defects in DNA repair mechanisms, leading to the accumulation of chromosomal abnormalities and gene mutations. This instability fosters heterogeneity within tumours, enhancing their adaptability and resistance to therapies.

Mechanisms contributing to genetic instability include errors in DNA replication, defective mismatch repair systems, and deficiencies in homologous recombination. The resulting genomic diversity within tumours poses significant challenges for treatment, as it facilitates the emergence of resistant cancer cell populations.

Advanced Concepts

Oncogenes and Their Activation

Oncogenes are mutated versions of proto-oncogenes that drive uncontrolled cell division when aberrantly activated. Proto-oncogenes normally regulate cell growth and differentiation, but mutations can convert them into constitutively active oncogenes. These mutations may involve point mutations, gene amplifications, or chromosomal translocations.

For example, the BCR-ABL fusion gene, resulting from a translocation between chromosomes 9 and 22, encodes a constitutively active tyrosine kinase. This abnormal kinase continuously signals for cell proliferation, leading to chronic myeloid leukemia (CML). Targeted therapies, such as imatinib, specifically inhibit the BCR-ABL kinase, exemplifying the therapeutic potential of targeting oncogenes.

Tumour Suppressor Genes and Loss of Function

Tumour suppressor genes safeguard against uncontrolled cell division by regulating the cell cycle, promoting DNA repair, and inducing apoptosis. Unlike oncogenes, which are dominant, mutations in tumour suppressor genes are typically recessive, requiring both alleles to be inactivated to disrupt their function. The "two-hit hypothesis" describes this inactivation process.

The RB1 gene, encoding the retinoblastoma protein (Rb), is a critical tumour suppressor involved in controlling the G1 to S phase transition. Loss of Rb function removes a key checkpoint, allowing cells with damaged DNA to progress through the cell cycle unchecked. Germline mutations in RB1 predispose individuals to retinoblastoma, a malignant eye tumour in children.

Signal Transduction Pathway Dysregulation

Signal transduction pathways, such as the PI3K/AKT/mTOR and Wnt/β-catenin pathways, play pivotal roles in regulating cell growth, metabolism, and survival. Dysregulation of these pathways can lead to persistent proliferative signals, resistance to apoptosis, and metabolic alterations conducive to tumour progression.

In the PI3K/AKT/mTOR pathway, mutations in PI3K or loss of the PTEN tumour suppressor result in hyperactivation of AKT, promoting cell survival and growth. Similarly, aberrant activation of the Wnt/β-catenin pathway enhances cell proliferation and stem cell-like properties, contributing to the aggressive nature of certain cancers.

Epigenetic Modifications in Cancer

Epigenetic modifications, including DNA methylation and histone acetylation, regulate gene expression without altering the DNA sequence. In cancer, epigenetic dysregulation can lead to the silencing of tumour suppressor genes or the activation of oncogenes. Hypermethylation of promoter regions is a common mechanism for silencing critical regulatory genes.

For instance, hypermethylation of the MLH1 gene promoter impairs DNA mismatch repair, increasing mutation rates and contributing to microsatellite instability in colorectal cancer. Epigenetic therapies, such as DNA methyltransferase inhibitors and histone deacetylase inhibitors, aim to restore normal gene expression patterns, offering promising avenues for cancer treatment.

Metastasis and the Epithelial-Mesenchymal Transition (EMT)

Metastasis, the spread of cancer cells from the primary tumour to distant sites, is a complex process involving multiple steps, including invasion, intravasation, circulation, extravasation, and colonization. The epithelial-mesenchymal transition (EMT) is a crucial event in metastasis, wherein epithelial cells acquire mesenchymal properties, enhancing their migratory and invasive capabilities.

During EMT, cells lose cell-cell adhesion mediated by E-cadherin and gain expression of mesenchymal markers like N-cadherin and vimentin. Transcription factors such as Snail, Slug, and Twist orchestrate these changes, facilitating the detachment and dissemination of cancer cells. Targeting EMT and metastatic pathways remains a significant focus in the development of anti-cancer therapies.

Genomic Instability and Cancer Stem Cells

Genomic instability not only contributes to genetic heterogeneity within tumours but also fosters the maintenance of cancer stem cells (CSCs). CSCs possess self-renewal capabilities and can differentiate into diverse cell types within the tumour, driving recurrence and resistance to conventional therapies.

Mutations that enhance DNA damage resistance and repair mechanisms allow CSCs to survive chemotherapy and radiation treatments. Targeting the specific pathways that sustain CSCs, such as the Notch, Hedgehog, and Wnt pathways, is being explored to prevent tumour relapse and improve treatment outcomes.

Immunoevasion Strategies of Tumours

Tumours employ various immunoevasion strategies to escape immune surveillance, enabling unchecked growth and metastasis. These strategies include the downregulation of major histocompatibility complex (MHC) molecules, secretion of immunosuppressive cytokines, and expression of checkpoint proteins like PD-L1.

By inhibiting immune cell activation and promoting an immunosuppressive tumour microenvironment, cancer cells evade detection and destruction by the immune system. Immunotherapies, such as checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4, aim to reinvigorate the immune response against cancer cells, demonstrating significant clinical efficacy in various malignancies.

Targeted Therapies and Precision Medicine

Advancements in genomics and molecular biology have paved the way for targeted therapies that specifically inhibit aberrant proteins driving cancer progression. Precision medicine involves tailoring treatment strategies based on the genetic and molecular profile of individual tumours, enhancing therapeutic efficacy and minimizing toxicity.

Examples of targeted therapies include tyrosine kinase inhibitors (e.g., imatinib for BCR-ABL-positive CML) and monoclonal antibodies (e.g., trastuzumab for HER2-positive breast cancer). Additionally, advancements in next-generation sequencing have facilitated the identification of actionable mutations, enabling personalized treatment regimens that improve patient outcomes.

Multidrug Resistance in Cancer Therapy

Multidrug resistance (MDR) poses a significant challenge in cancer treatment, as cancer cells develop mechanisms to evade the cytotoxic effects of chemotherapy. MDR can arise from various factors, including the overexpression of drug efflux pumps like P-glycoprotein, alterations in drug targets, enhanced DNA repair, and evasion of apoptosis.

Efforts to overcome MDR involve the use of combination therapies that target multiple pathways simultaneously, the development of inhibitors for drug efflux pumps, and the implementation of novel drug delivery systems that bypass resistance mechanisms. Understanding the molecular basis of MDR is essential for devising effective therapeutic strategies to combat resistant cancers.

Microenvironmental Influences on Tumour Growth

The tumour microenvironment (TME) comprises various non-cancerous cells, extracellular matrix components, and signaling molecules that interact with cancer cells, influencing their growth and behavior. Components of the TME include fibroblasts, immune cells, endothelial cells, and cytokines, which collectively modulate tumour progression, angiogenesis, and metastasis.

Interactions within the TME can promote cancer cell survival, enhance invasive properties, and facilitate resistance to therapies. Targeting the TME, such as inhibiting angiogenesis or modulating immune cell activity, represents a promising approach in cancer treatment, complementing strategies aimed directly at cancer cells.

Comparison Table

Aspect Normal Cell Division Uncontrolled Cell Division
Regulation Strictly controlled by cell cycle checkpoints Disrupted regulation due to genetic mutations
Growth Signals Respond to external growth factors Independent of external signals; constitutively active
Apoptosis Activated in response to DNA damage Inhibited or evaded, allowing survival of damaged cells
Genomic Stability High fidelity DNA replication and repair Genetic instability with accumulation of mutations
Cell Adhesion Maintain proper cell-cell adhesion Altered adhesion, facilitating invasion and metastasis

Summary and Key Takeaways

  • Uncontrolled cell division leads to tumour formation by disrupting normal regulatory mechanisms.
  • Oncogenes and tumor suppressor genes play pivotal roles in regulating cell proliferation and preventing malignancies.
  • Genetic and epigenetic alterations contribute to the accumulation of mutations and genomic instability in cancer cells.
  • Advanced concepts include signal transduction dysregulation, cancer stem cells, and tumour microenvironment influences.
  • Understanding these mechanisms is crucial for developing targeted therapies and improving cancer treatment strategies.

Coming Soon!

coming soon
Examiner Tip
star

Tips

To excel in exams, create mnemonics for remembering key oncogenes and tumour suppressor genes, such as "RATS MAKE PIES" for RAS, MYC, and P53. Use diagrams to visualize signal transduction pathways and their dysregulations in cancer. Practice differentiating normal and uncontrolled cell division scenarios with comparison tables. Additionally, stay updated with recent cancer therapies to understand practical applications of theoretical concepts.

Did You Know
star

Did You Know

Did you know that the discovery of the p53 protein, often dubbed the "guardian of the genome," has been pivotal in understanding cancer mechanisms? Additionally, some tumours can manipulate their surrounding environment to promote blood vessel growth, a process known as angiogenesis, which is crucial for their survival and expansion. Moreover, research has revealed that certain cancers can develop resistance to therapies by activating alternative signaling pathways, making treatment more challenging.

Common Mistakes
star

Common Mistakes

One common mistake is confusing oncogenes with tumour suppressor genes; remember that oncogenes promote cell division when mutated, while tumour suppressors inhibit it. Another error students make is overlooking the role of apoptosis in preventing cancer; failing to recognize how the inhibition of programmed cell death contributes to uncontrolled growth. Additionally, students often misinterpret the distinction between genetic and epigenetic changes in cancer development.

FAQ

What is the primary difference between oncogenes and tumour suppressor genes?
Oncogenes are mutated genes that promote uncontrolled cell division, whereas tumour suppressor genes inhibit cell division and prevent tumour formation.
How does the p53 protein contribute to preventing cancer?
p53 induces cell cycle arrest or apoptosis in response to DNA damage, preventing the propagation of genetically abnormal cells.
What role does angiogenesis play in tumour growth?
Angiogenesis provides tumours with the necessary blood supply for nutrients and oxygen, enabling them to grow beyond a limited size and facilitating metastasis.
Why is genetic instability a challenge in cancer treatment?
Genetic instability leads to tumour heterogeneity, making it difficult to target all cancer cells effectively and increasing the likelihood of treatment resistance.
What are some common targeted therapies used in cancer treatment?
Examples include tyrosine kinase inhibitors like imatinib for CML and monoclonal antibodies such as trastuzumab for HER2-positive breast cancer.
Download PDF
Get PDF
Download PDF
PDF
Share
Share
Explore
Explore
How would you like to practise?
close