Molecular Mechanisms of Cancer: The Role of Oncogenes and Tumor Suppressor Genes in Tumorigenesis

Introduction

Cancer is a complex disease characterized by uncontrolled cell division due to genetic and molecular abnormalities. Two crucial classes of genes—oncogenes and tumor suppressor genes—play a vital role in the development and progression of cancer. Understanding the molecular basis of cancer helps in targeted therapies, early diagnosis, and personalized medicine.


Role of oncogenes in cancer development,
Tumor suppressor genes and cancer prevention,
How mutations cause cancer growth,
Molecular genetics behind cancer formation,
Cancer progression and genetic mutations.

The Molecular Basis of Cancer

Cancer arises due to mutations in genes that regulate cell cycle control, apoptosis, DNA repair, and signaling pathways. Mutations in oncogenes and tumor suppressor genes disrupt normal cell functions, leading to unregulated growth and tumor formation.

Oncogenes: The Drivers of Cancer

Definition of Oncogenes

Oncogenes are mutated or overexpressed genes that promote excessive cell proliferation and survival. They are derived from normal cellular genes called proto-oncogenes, which regulate normal cell growth and differentiation.

Activation of Oncogenes

Proto-oncogenes become oncogenes due to mutations, gene amplification, or chromosomal translocations. Some key mechanisms include:

  • Point Mutations: A single nucleotide change leading to hyperactive proteins (e.g., RAS gene mutation).
  • Gene Amplification: Increased copy numbers of proto-oncogenes resulting in overexpression (e.g., HER2 in breast cancer).
  • Chromosomal Translocation: Exchange of genetic material leading to novel fusion proteins (e.g., BCR-ABL in chronic myeloid leukemia).

Examples of Oncogenes

  1. RAS Family: Mutations in RAS genes (KRAS, HRAS, NRAS) lead to persistent cell signaling, resulting in unchecked proliferation.
  2. MYC: A transcription factor that promotes cell growth and division.
  3. HER2/Neu: Overexpressed in breast and gastric cancers, leading to aggressive tumor growth.
  4. BCR-ABL: A fusion oncogene in chronic myeloid leukemia that continuously activates cell division.

Tumor Suppressor Genes: The Guardians Against Cancer

Definition of Tumor Suppressor Genes

Tumor suppressor genes regulate cell cycle progression, DNA repair, and apoptosis. Loss of function in these genes leads to uncontrolled cell proliferation and tumorigenesis.

Mechanisms of Tumor Suppressor Gene Inactivation

  • Point Mutations: Inactivating mutations (e.g., TP53 mutations in many cancers).
  • Deletion: Loss of entire gene regions leading to loss of function.
  • Epigenetic Silencing: DNA methylation and histone modifications that silence gene expression.

Examples of Tumor Suppressor Genes

  1. TP53 (p53 Protein): Known as the “guardian of the genome,” TP53 regulates apoptosis and DNA repair. Mutations in TP53 are found in over 50% of human cancers.
  2. RB1 (Retinoblastoma Protein): Controls cell cycle progression by inhibiting excessive cell proliferation.
  3. BRCA1 and BRCA2: Involved in DNA repair; mutations increase the risk of breast and ovarian cancers.
  4. PTEN: A phosphatase that regulates cell survival pathways; mutations contribute to multiple cancers.

The Interplay Between Oncogenes and Tumor Suppressor Genes

  • Cancer results from a balance shift between oncogenic activation and tumor suppressor gene inactivation.
  • Multiple genetic and epigenetic events drive tumor progression, leading to metastasis and therapy resistance.
  • Two-Hit Hypothesis (Knudson’s Theory): Suggests that both alleles of a tumor suppressor gene must be inactivated for cancer development.

Emerging Targeted Therapies Based on Molecular Understanding

Targeting Oncogenes

  • Tyrosine Kinase Inhibitors (TKIs): Imatinib (Gleevec) for BCR-ABL fusion protein in chronic myeloid leukemia.
  • HER2 Inhibitors: Trastuzumab (Herceptin) for HER2-positive breast cancer.
  • RAS Pathway Inhibitors: Targeting mutant KRAS-driven cancers.

Restoring Tumor Suppressor Function

  • Gene Therapy: Introducing functional TP53 into cancer cells.
  • Epigenetic Drugs: DNA methylation inhibitors to reactivate silenced tumor suppressor genes.

Prevention and Future Directions

  • Early Screening: Detecting mutations in oncogenes and tumor suppressor genes for personalized medicine.
  • Lifestyle Modifications: Reducing carcinogen exposure (e.g., tobacco, UV radiation) to prevent mutations.
  • Immunotherapy Advances: Enhancing immune response against tumor cells using checkpoint inhibitors (e.g., PD-1 inhibitors).

Relevant Website URL Links

Further Reading

Conclusion

Understanding the molecular basis of cancer through oncogenes and tumor suppressor genes has revolutionized cancer diagnosis, prognosis, and treatment strategies. Advances in genetic research and targeted therapies hold promise for improved cancer management and personalized medicine.



MCQs on “Molecular Basis of Cancer: Role of Oncogenes and Tumor Suppressor Genes”

1. What is the primary cause of cancer at the molecular level?

A) Viral infections
B) Genetic mutations
C) Poor diet
D) Bacterial infections

Answer: B) Genetic mutations
Explanation: Cancer occurs due to mutations in genes that regulate cell growth and division, such as oncogenes and tumor suppressor genes.


2. Which of the following genes is classified as an oncogene?

A) p53
B) BRCA1
C) MYC
D) RB1

Answer: C) MYC
Explanation: MYC is an oncogene that promotes cell proliferation and survival. Mutations or overexpression of MYC contribute to cancer progression.


3. What role do tumor suppressor genes play in cancer?

A) They promote cell division
B) They repair damaged DNA or induce apoptosis
C) They prevent mutations from occurring
D) They inhibit the immune system

Answer: B) They repair damaged DNA or induce apoptosis
Explanation: Tumor suppressor genes, such as p53 and RB1, help regulate cell cycle checkpoints, DNA repair, and apoptosis. Their inactivation leads to uncontrolled cell growth.


4. Which of the following tumor suppressor genes is known as the “guardian of the genome”?

A) p53
B) HER2
C) RAS
D) VEGF

Answer: A) p53
Explanation: p53 plays a crucial role in preventing genetic mutations by triggering DNA repair, cell cycle arrest, or apoptosis in response to cellular stress.


5. Mutations in which gene are most commonly associated with hereditary breast and ovarian cancer?

A) BRCA1 and BRCA2
B) APC
C) Rb
D) KRAS

Answer: A) BRCA1 and BRCA2
Explanation: Mutations in BRCA1 and BRCA2 impair DNA repair mechanisms, increasing the risk of breast and ovarian cancer.


6. Which of the following is an example of a proto-oncogene that can become an oncogene when mutated?

A) TP53
B) RAS
C) BRCA1
D) PTEN

Answer: B) RAS
Explanation: RAS is a proto-oncogene that, when mutated, remains in an active state, continuously promoting cell division and contributing to cancer.


7. How do oncogenes contribute to cancer development?

A) They suppress tumor growth
B) They prevent DNA replication
C) They lead to uncontrolled cell proliferation
D) They repair damaged DNA

Answer: C) They lead to uncontrolled cell proliferation
Explanation: Oncogenes drive continuous cell growth and division, bypassing normal regulatory mechanisms.


8. Which of the following is NOT a mechanism of tumor suppressor gene inactivation?

A) Gene deletion
B) Loss of heterozygosity
C) Chromosomal translocation
D) DNA amplification

Answer: D) DNA amplification
Explanation: Tumor suppressor genes are inactivated through deletion, mutation, or epigenetic silencing. DNA amplification usually leads to oncogene activation.


9. The two-hit hypothesis applies to which type of genes?

A) Oncogenes
B) Tumor suppressor genes
C) DNA repair genes
D) Growth factor genes

Answer: B) Tumor suppressor genes
Explanation: The two-hit hypothesis states that both alleles of a tumor suppressor gene must be inactivated to contribute to cancer development.


10. Which oncogene is frequently mutated in human cancers and involved in signal transduction?

A) RAS
B) TP53
C) APC
D) BRCA2

Answer: A) RAS
Explanation: Mutations in the RAS gene lead to continuous activation of signaling pathways that drive cell proliferation.


11. Which type of cancer is commonly associated with APC gene mutations?

A) Lung cancer
B) Colorectal cancer
C) Breast cancer
D) Melanoma

Answer: B) Colorectal cancer
Explanation: APC is a tumor suppressor gene that regulates Wnt signaling, and mutations in APC are commonly seen in colorectal cancer.


12. What is the primary function of the RB1 gene?

A) Activating cell division
B) Inhibiting cell cycle progression
C) Inducing angiogenesis
D) Promoting metastasis

Answer: B) Inhibiting cell cycle progression
Explanation: The RB1 protein (retinoblastoma protein) prevents uncontrolled cell proliferation by regulating the G1/S transition in the cell cycle.


13. Which pathway is most commonly activated by oncogenic RAS mutations?

A) Wnt signaling
B) MAPK/ERK pathway
C) p53 pathway
D) TGF-β signaling

Answer: B) MAPK/ERK pathway
Explanation: RAS mutations activate the MAPK/ERK pathway, leading to increased cell division and survival.


14. How do tumor suppressor genes become inactivated in cancer?

A) Activation mutations
B) Deletions, mutations, or epigenetic silencing
C) Overexpression
D) Fusion with oncogenes

Answer: B) Deletions, mutations, or epigenetic silencing
Explanation: Loss of function in tumor suppressor genes can result from deletions, mutations, or epigenetic modifications like DNA methylation.


15. Which virus is most strongly associated with cervical cancer?

A) Hepatitis B virus
B) Epstein-Barr virus
C) Human papillomavirus (HPV)
D) Human immunodeficiency virus (HIV)

Answer: C) Human papillomavirus (HPV)
Explanation: HPV produces E6 and E7 proteins that inactivate tumor suppressors p53 and RB1, leading to cervical cancer.


16. Which process allows cancer cells to evade programmed cell death (apoptosis)?

A) Overexpression of p53
B) Activation of the Bcl-2 family proteins
C) Increased production of reactive oxygen species
D) Downregulation of RAS

Answer: B) Activation of the Bcl-2 family proteins
Explanation: Bcl-2 is an anti-apoptotic protein that prevents programmed cell death, allowing cancer cells to survive despite stress and damage.


17. The Philadelphia chromosome (BCR-ABL fusion) is associated with which type of cancer?

A) Breast cancer
B) Chronic myeloid leukemia (CML)
C) Lung cancer
D) Glioblastoma

Answer: B) Chronic myeloid leukemia (CML)
Explanation: The BCR-ABL fusion gene, formed by chromosomal translocation (t9;22), leads to continuous activation of tyrosine kinase signaling, driving leukemia.


18. Which gene mutation is most commonly found in human cancers?

A) BRCA2
B) APC
C) p53
D) Rb

Answer: C) p53
Explanation: Mutations in the TP53 gene are found in over 50% of human cancers, leading to loss of cell cycle arrest and apoptosis regulation.


19. What is the function of telomerase in cancer cells?

A) Inducing apoptosis
B) Limiting cell division
C) Maintaining telomere length and enabling unlimited replication
D) Promoting immune response

Answer: C) Maintaining telomere length and enabling unlimited replication
Explanation: Cancer cells reactivate telomerase, which prevents telomere shortening and allows them to divide indefinitely.


20. Which of the following is a hallmark of cancer?

A) Increased DNA repair efficiency
B) Evasion of growth suppressors
C) Reduced angiogenesis
D) Limited replicative potential

Answer: B) Evasion of growth suppressors
Explanation: Cancer cells avoid normal regulatory mechanisms by inactivating tumor suppressor genes and promoting unchecked growth.


21. Loss of function mutations in which gene lead to Li-Fraumeni syndrome?

A) RB1
B) BRCA1
C) TP53
D) PTEN

Answer: C) TP53
Explanation: Li-Fraumeni syndrome is caused by inherited p53 mutations, leading to a high risk of various cancers at an early age.


22. How does angiogenesis contribute to tumor progression?

A) It inhibits metastasis
B) It supplies oxygen and nutrients to the tumor
C) It prevents immune system attack
D) It repairs damaged DNA

Answer: B) It supplies oxygen and nutrients to the tumor
Explanation: Tumors induce blood vessel formation (angiogenesis) by secreting factors like VEGF, promoting their growth and survival.


23. Which of the following genes acts as a tumor suppressor?

A) KRAS
B) HER2
C) TP53
D) BCR-ABL

Answer: C) TP53
Explanation: p53 regulates the cell cycle, promotes DNA repair, and induces apoptosis when necessary, preventing uncontrolled cell growth.


24. What is the role of E-cadherin in cancer?

A) It promotes metastasis
B) It enhances cell adhesion and prevents invasion
C) It activates oncogenes
D) It induces angiogenesis

Answer: B) It enhances cell adhesion and prevents invasion
Explanation: Loss of E-cadherin, a cell adhesion molecule, allows cancer cells to detach and metastasize.


25. Which cancer treatment strategy targets oncogenic tyrosine kinases like BCR-ABL?

A) Chemotherapy
B) Immunotherapy
C) Tyrosine kinase inhibitors (TKIs)
D) Hormone therapy

Answer: C) Tyrosine kinase inhibitors (TKIs)
Explanation: Drugs like Imatinib (Gleevec) target oncogenic tyrosine kinases, effectively treating cancers such as CML.


26. How do cancer cells achieve immune evasion?

A) Overexpression of PD-L1
B) Increased apoptosis
C) Enhanced DNA repair mechanisms
D) Reduced telomerase activity

Answer: A) Overexpression of PD-L1
Explanation: PD-L1 binds to PD-1 on immune cells, suppressing their activity and allowing cancer cells to escape immune destruction.


27. How do mutations in PTEN contribute to cancer?

A) By increasing cell adhesion
B) By promoting angiogenesis
C) By activating the PI3K-AKT pathway
D) By inducing apoptosis

Answer: C) By activating the PI3K-AKT pathway
Explanation: PTEN negatively regulates the PI3K-AKT pathway, which promotes cell survival and growth. Loss of PTEN leads to unchecked proliferation.


28. Which signaling pathway is frequently involved in colon cancer?

A) Notch signaling
B) Wnt/β-catenin pathway
C) MAPK pathway
D) PI3K-AKT pathway

Answer: B) Wnt/β-catenin pathway
Explanation: Mutations in the APC gene disrupt Wnt signaling, leading to abnormal β-catenin accumulation and colorectal cancer progression.


29. What is the role of BRCA1 in normal cells?

A) Regulating cell cycle progression
B) Repairing double-strand DNA breaks
C) Suppressing immune response
D) Stimulating angiogenesis

Answer: B) Repairing double-strand DNA breaks
Explanation: BRCA1 is involved in homologous recombination, a key DNA repair mechanism. Mutations in BRCA1 increase susceptibility to breast and ovarian cancer.


30. Which of the following best describes the Warburg effect in cancer cells?

A) Increased oxidative phosphorylation
B) Increased glycolysis even in the presence of oxygen
C) Reduced glucose uptake
D) Enhanced fatty acid oxidation

Answer: B) Increased glycolysis even in the presence of oxygen
Explanation: The Warburg effect describes how cancer cells preferentially use aerobic glycolysis for energy production, even when oxygen is available, leading to increased glucose consumption.



LEAVE A REPLY

Please enter your comment!
Please enter your name here