Molecular Cell Biology. Prof. D. Karunagaran. Department of Biotechnology. Indian Institute of Technology Madras

Similar documents
mirna Dr. S Hosseini-Asl

R. Piazza (MD, PhD), Dept. of Medicine and Surgery, University of Milano-Bicocca EPIGENETICS

Epigenetics. Lyle Armstrong. UJ Taylor & Francis Group. f'ci Garland Science NEW YORK AND LONDON

Genetics and Genomics in Medicine Chapter 6 Questions

Jayanti Tokas 1, Puneet Tokas 2, Shailini Jain 3 and Hariom Yadav 3

Epigenetics Armstrong_Prelims.indd 1 04/11/2013 3:28 pm

Histones modifications and variants

Ch. 18 Regulation of Gene Expression

Epigenomics. Ivana de la Serna Block Health Science

Biochemical Determinants Governing Redox Regulated Changes in Gene Expression and Chromatin Structure

Regulation of Gene Expression in Eukaryotes

Gene Expression DNA RNA. Protein. Metabolites, stress, environment

Lecture 8. Eukaryotic gene regulation: post translational modifications of histones

Stem Cell Epigenetics

Transcriptional control in Eukaryotes: (chapter 13 pp276) Chromatin structure affects gene expression. Chromatin Array of nuc

Session 2: Biomarkers of epigenetic changes and their applicability to genetic toxicology

Eukaryotic transcription (III)

Epigenetics q&more 01.11

Epigenetic Mechanisms

Eukaryotic Gene Regulation

Are you the way you are because of the

Introduction to Genetics

Epigenetics in evolution and disease

Not IN Our Genes - A Different Kind of Inheritance.! Christopher Phiel, Ph.D. University of Colorado Denver Mini-STEM School February 4, 2014

Peter A. Jones* and Stephen B. Baylin

Epigenetics DNA methylation. Biosciences 741: Genomics Fall, 2013 Week 13. DNA Methylation

Epigenetic Inheritance

EPIGENOMICS PROFILING SERVICES

Chromatin-Based Regulation of Gene Expression

DNA methylation & demethylation

Transcription Regulation And Gene Expression in Eukaryotes Cycle G2 (lecture 13709) FS 2014 P Matthias & RG Clerc

The Biology and Genetics of Cells and Organisms The Biology of Cancer

Overview: Conducting the Genetic Orchestra Prokaryotes and eukaryotes alter gene expression in response to their changing environment

Epigenetics and Human Disease

The silence of the genes: clinical applications of (colorectal) cancer epigenetics

I) Development: tissue differentiation and timing II) Whole Chromosome Regulation

Changes in DNA methylation and histone modification during epigenetic transitions

Epigenetics and Colorectal Cancer Pathogenesis

Epigenetics and Environmental Health A Step-by-Step Tutorial

Introduction to Cancer Biology

MicroRNA in Cancer Karen Dybkær 2013

FOLLICULAR LYMPHOMA- ILLUMINA METHYLATION. Jude Fitzgibbon

Dominic J Smiraglia, PhD Department of Cancer Genetics. DNA methylation in prostate cancer

Chapter 19 Eukaryotic Genomes

Epigenetics: Basic Principals and role in health and disease

Epigenetics & cancer. Present by : Sanaz Zebardast Under supervision : Dr. Gheibi. 31 December 2016

MicroRNA dysregulation in cancer. Systems Plant Microbiology Hyun-Hee Lee

Epigenetics ~from mechanism to therapy~ Literature seminar January 31, 2012 Soichi Ito (B4)

DNA Methylation and Cancer

MicroRNA and Male Infertility: A Potential for Diagnosis

Genetics. Instructor: Dr. Jihad Abdallah Transcription of DNA

Imprinting. Joyce Ohm Cancer Genetics and Genomics CGP-L2-319 x8821

Epigenetic gene silencing in cancer a mechanism for early oncogenic pathway addiction?

Abstract. KEY WORDS: epigenetics, DNA methylation, histone modification, one-carbon metabolism, stem cell aging, anti-aging medicine

Repressive Transcription

Transformation of Normal HMECs (Human Mammary Epithelial Cells) into Metastatic Breast Cancer Cells: Introduction - The Broad Picture:

Epigenetics: A historical overview Dr. Robin Holliday

LESSON 3.2 WORKBOOK. How do normal cells become cancer cells? Workbook Lesson 3.2

Prokaryotes and eukaryotes alter gene expression in response to their changing environment

Molecular Biology (BIOL 4320) Exam #2 May 3, 2004

Genetics and Genomics in Medicine Chapter 6. Questions & Answers

DNA methylation: a potential clinical biomarker for the detection of human cancers

Fragile X Syndrome. Genetics, Epigenetics & the Role of Unprogrammed Events in the expression of a Phenotype

Epigenetics: The Future of Psychology & Neuroscience. Richard E. Brown Psychology Department Dalhousie University Halifax, NS, B3H 4J1

Epigenetic Aberrations in Cholangiocarcinoma: Potential Biomarkers and Promising Target for Novel Therapeutic Strategies

Epigenetic mechanisms behind cellular sensitivity to DNA damage

Removal of Shelterin Reveals the Telomere End-Protection Problem

DSB. Double-Strand Breaks causate da radiazioni stress ossidativo farmaci

Removal of Shelterin Reveals the Telomere End-Protection Problem

Cell Quality Control. Peter Takizawa Department of Cell Biology

Cancer and Gene Regulation

Epigenetics and Toxicology

BIO360 Fall 2013 Quiz 1

Lecture 10. Eukaryotic gene regulation: chromatin remodelling

The mutations that drive cancer. Paul Edwards. Department of Pathology and Cancer Research UK Cambridge Institute, University of Cambridge

Cancer Letters xxx (2012) xxx xxx. Contents lists available at SciVerse ScienceDirect. Cancer Letters

Development of Carcinoma Pathways

HGD 5502 EPIGENETICS. Dr. Abhi Veerakumarasivam (2011)

STEM CELL GENETICS AND GENOMICS

p53 cooperates with DNA methylation and a suicidal interferon response to maintain epigenetic silencing of repeats and noncoding RNAs

A Genetic Program for Embryonic Development

Mag. a Angelika Pointner Dep. for Nutritional Sciences University of Vienna. Einfluss pflanzlicher Substanzen auf Telomere und Epigenetik

Aberrant Promoter CpG Methylation is a Mechanism for Lack of Hypoxic Induction of

4/20/2016. Objectives. Epigenetic Definitions. Gene Expression. More Questions. Questions to Consider

SUPPLEMENTARY INFORMATION

Cover Page. The handle holds various files of this Leiden University dissertation.

An Overview of the Molecular Basis of Epigenetics

Dr Kavita Raj Consultant Haematologist Guys and St Thomas Hospital

MicroRNA expression profiling and functional analysis in prostate cancer. Marco Folini s.c. Ricerca Traslazionale DOSL

4/8/2016. Objectives. Epigenetic Definitions. Gene Expression. More Questions. Epigentics. Questions to Consider

Transcription and chromatin. General Transcription Factors + Promoter-specific factors + Co-activators

Section D: The Molecular Biology of Cancer

Epigenetic therapy in urologic cancers: an update on clinical trials

MicroRNAs, RNA Modifications, RNA Editing. Bora E. Baysal MD, PhD Oncology for Scientists Lecture Tue, Oct 17, 2017, 3:30 PM - 5:00 PM

MCB 102 Third Exam Spring 2015

DNA Methylation and Demethylation as Targets for Anticancer Therapy

Chapter 11 How Genes Are Controlled

Review. Ageing 2: Cancer! Review: Mutations. Mutations 2/14/11. The Raw Material for Evolution. The Double Edged Sword

Gene Regulation Part 2

EPIGENETICS AND MENTAL HEALTH. William J. Walsh, Ph.D.

Transcription:

Molecular Cell Biology Prof. D. Karunagaran Department of Biotechnology Indian Institute of Technology Madras Module-9 Molecular Basis of Cancer, Oncogenes and Tumor Suppressor Genes Lecture 6 Epigenetics and Cancer

Introduction In the human genome we have about 20-30,000 genes Tissue specific gene expression is known to contribute for the identity of that tissue Earlier we learnt that Mutations, deletions, etc lead to loss of expression of tumor suppressor genes in a tumor Epigenetic silencing of tumor suppressor genes is another way, perhaps a more prevalent one, that accounts for the loss of expression of tumor suppressor genes in cancer What is epigenetics? Genetics could not explain all the differences between phenotype and genotype in some of the processes such as differentiation. For instance genetics alone cannot account for drastic changes observed between undifferentiated and differentiated cells of the same tissue given the idea that these cells have the same set of genes or genetic information. Hence it was thought that the undifferentiated cells undergo a crisis first that determines their fate This crisis was not dependent on their genes and so it was thought that this happens in addition to genes (in Greek it is epigenetic) What is epigenetics? Conrad Waddington in 1939 first coined the term epigenetics (literally meaning above genetics ) Epigenetics means the study of mitotically and/or meiotically heritable changes in gene expression that occur without a change in DNA sequence mediated mainly through changes in DNA methylation and chromatin structure. Human tumors exhibit an overall loss of DNA methylation, but also show specific patterns of hyper methylation at certain promoters

DNA packaging We have learnt about chromosomes, chromatin and nucleosomes. DNA packaging involves nucleosomes characterized by DNA chains (~146bp) wrapping around histone octamers. Histones are small (11 21 kda) basic proteins that bind non-covalently with acidic DNA to form nucleosomes. Octamers are formed by H2A, H2B, H3 and H4 dimers DNA methylation and Histone modifications Open and closed chromatin Portions of DNA bound with histones and other proteins should be exposed for facilitating transcription factor binding. This influences the gene expression profile of any given cell type Nucleosomes can be packaged into compact aggregates silencing transcription and thus constituting a closed chromatin In contrast nucleosomes can also be arranged leaving some gaps for transcription factors to bind constituting an open chromatin Thus the chromatin is said to be bivalent. Post-translational modification of key amino acid residues of the histones is important in this connection These modifications constitute the so called histone code for the regulation of gene expression

Histone modifications Histones are not just only involved in DNA-packaging They undergo several modifications important for gene regulation Covalent modification of histones includes acetylation of lysines, methylation of lysines and arginines, Phosphorylations of serines and threonines, ADP-ribosylation of glutamic acids, and ubiquitination and sumolyation of lysine residues The status of acetylation and methylation of specific lysine residues contained within the tails of nucleosomal core histones is known to have a crucial role in regulating chromatin structure and gene expression Acetylation of some residues, such as lysine 9 of histone H3 (H3K9acetyl), by histone acetylases (HATs) usually signifies transcriptionally activeregions Deacetylation at these sites, mediated by histone deactylases (HDACs) usually signifies transcriptional repression. Histone tail methylation can be activating or inactivating transcription depending on the sites. H3K4 methylation denotes transcriptionally active areas while H3K9 methylation or H3K27 methylation suggests transcriptionally repressed regions. Histone methyltransferases place these methylation marks (Writers of the code) Histone demethylases remove the marks (Erasers of the code) Methylation Well known epigenetic modification in humans is the cytosine methylation of DNA within the dinucleotide CpG. CpG is used to refer to both methylated and unmethylated dinucleotides p refers to the phosphate moiety that connects deoxycytidine and deoxyguanosine CpGs occur in the genome at a lower frequency because methylated cytosines can spontaneously deaminate to form thymine

DNA repair machinery does not recognize this leading to the accumulation of C-T mutations during evolution Thus "99%" of the genome is CpG depleted and 1% is composed of discrete regions that have a high (G+C) and CpG content DNA methylation occurs along with other epigenetic modifications. It is connected closely with the formation of nuclease-resistant chromatin Methyl-CpG-binding proteins and DNA methyltransferases (DNMTs) are associated with histone deacetylases and histone methyl transferases. CpG-rich regions are known as CpG islands Regions of comparatively low CpG density close to CpG islands are called CpG island shores In cancer cells, the transcriptional silencing of tumour-suppressor genes occurs by CpG-island-promoter hypermethylation Deamination and Methylation

Cytosine upon oxidative deamination changes into uracil. This will be considered a mutation and hence will be removed by the DNA repair system and that is why we have Thymine but not uracil in DNA Methylated cytosine upon oxidative deamination changes into thymine Source of methylation

CpG sites are concentrated either in CpG islands, located in approximately 60% of human gene promoters, or in regions of large repetitive sequences (for example, centromeres and retrotransposon elements). Unmethylated clusters of CpG pairs are located in tissue specific genes and in essential housekeeping genes (involved in routine maintenance roles and are expressed in most tissues). There are specific proteins that bind to unmethylated CpGs and initiate gene transcription. Methyl groups are acquired through the diet and are donated to DNA through the folate and methionine pathways with S-adenosyl methionine being the donor of methyl groups that is covalently attached to the cytosine. Low dietary levels of folate, methionine or selenium can affect methylation Such defects are associated with neural tube defects, cancer and atherosclerosis Effects of Methylation Thus imbalances in dietary nutrients can cause hypomethylation This may lead to improper gene expression and genetic instability (chromosome rearrangements). A common polymorphism in the methylene tetrahydrofolate reductase (MTHFR) gene (MTHFR 677CT) increases the risk of breast cancer up to 3- fold in premenopausal women Repetitive genomic sequences are heavily methylated. DNA methylation protects chromosomal integrity, by preventing chromosomal instability, translocations and gene disruption Genomic imprinting requires DNA hyper methylation at one of the two parental alleles of a gene in order to establish mono allelic expression Loss of imprinting is a mechanism of gene activation in some types of cancer Gene-dosage reduction is observed in X-chromosome inactivation in females. DNA methylation is needed for the germ line-specific expression of genes Methylation is a mechanism for silencing tissue-specific genes in cell types in which they should not be expressed

Changes in DNA methylation in cancer There is an overall decrease in the levels of DNA methylation in cancers The loss in methylation is evident within repeat sequences, and promoters of selected genes, and in the pericentromeric regions of chromosomes These changes are linked with abnormal gene expression and chromosomal instability. Promoter DNA hypermethylation is observed more frequently in cancers than mutations This is important since it is a known mechanism for loss of tumor suppressor gene function The genes affected include those involved in cell adhesion cadherin DNA repair MLH1 Cell migration TIMPs Differentiation TGF beta receptor Hypermethylation in human cancers Cancer Hyper methylated gene Breast, Ovary BRCA1 Cervix p16 Kidney TIMP-3 Lung P16, p73 Prostate BRCA2 Pancreas APC Uterus hmlh1

Interactions between DNA methylation and chromatin changes H3K9 methylation and the histone methyltransferases are required for DNA methylation in lower organisms such as Arabidopsis and Neurospora. Polycomb group of proteins, which target another key gene repression mark to nucleosomes, H3K27me, has been implicated in the targeting and maintenance of DNA methylation. Methyl cytosine binding proteins (MBPs), and the protein complexes in which they reside, can bind to methylated CpG sites to help relay a silencing signal. These complexes contain histone deacetylases ( HDACs), which catalyze the deacetylation of key amino acid residues, such as H3K9, that are highly characteristic of transcriptionally silent regions of DNA The DNMTs themselves also interact with HDACs This helps to target these enzymes to sites of DNA methylation. Changes in cancer include increases in the levels and activities of the DNA methylation catalyzing enzymes or the proteins in complexes that modulate the enzymes that catalyze transcriptional repression histone modifications or altered levels of the repressive histone marks loss of acetylation at H4K16 and increased levels of H4K20 acetylation Interactions between DNA methylation and chromatin changes DNA methylation at gene promoters can result in gene silencing Histone modifications associated with active gene transcription such as acetylation of H3K9 and H4K16 decrease Histone modifications associated with transcriptionally repressive chromatin such as H3K9me2 and me3, and H3K27me3 increase The enzymes involved in the repressive marks increase in their activities. H3K27me3 histone modification is an important indication in human embryonic stem cells for genes that would become hypermethylated in adult cancers. Additionally H3K9 me2 and the H3K27 me3 mark may be involved

Gene silencing and tumor progression Although the loss and gain of methylation can occur at any of the multistages of cancer development, most often these happen at the initial stages For instance loss of a tumor suppressor gene may give a survival advantage for a preneoplastic cell or progenitor or stem cells Such a change may also confer the ability withstand adverse circumstances such as chronic inflammation, ROS, hypoxia etc and still make a progress towards carcinogenesis Cells encountering DNA damage would normally undergo apoptosis but if these changes in methylation occur they may survive and expand and select for mutations or other changes that favor tumor progression p16 gene is a frequent target for early methylation in breast cancer and nonsmall cell lung cancer,\. Histologically normal mammary epithelium from some women without malignancy can harbor p16 promoter hypermethylation Experimentally, early loss of p16 in mammary epithelial cells precedes genomic and epigenetic instability. In colon cancer aberrant crypt foci show DNA hypermethylation of a family of genes, the SFRPs, (secreted frizzled receptor proteins) that antagonize Wnt interaction with its receptors. If this hypermethylation persists throughout colon tumor progression it may drive the Wnt pathway further in collusion with the downstream mutations Translational aspects of epigenetic changes Understanding on the epigenetic changes in cancer may provide potential biomarkers for use in cancer risk assessment, early diagnosis, and prognosis assessment. H3 and H4 modifications correlate with aggressiveness of prostate cancer. In several cancer aggressive behavior of cells correlate with increases in levels of DNA methyltransferases and histone methyltransferases such as EZH2 for H3K27 methylation, and other PcG (polycomb group) gene silencing constituents. Reversal of DNA hypermethylation and aberrant gene silencing are promising strategies to combat cancer

Development of molecular markers One idea is to use promoter hypermethylation sequences as a molecular signature (as a biomarker) for each type of cancer. Some studies with sputum DNA from patients at high risk for lung cancer have shown that invasive tumors may be predicted, more than a year before clinical detection of cancer. Abnormal methylation markers in sputum were useful for predicting recurrence of the disease in patients with surgically resected early stage lung cancers. p16 gene in DNA from lung cancers predicts poor outcome. In normal colon tissues gene promoter methylation increases with age and parallels the risk of cancer. Metastatic potential can be predicted on the basis of the E-cadherin promoter methylation in breast and oral cancers O 6 -MGMT is a gene that encodes for a protein that mediates removal of bulky alkylation adducts from guanosines. Silencing of O 6 -MGMT gene is observed in many cancers Consequently these cancer cells cannot efficiently repair alkylation damage Such cancer cells are sensitive to alkylating agents such as temozolomide. Thus brain tumors harboring O6-MGMT respond better to temozolomide Strategies for cancer prevention and therapy Gene silencing by epigenetic alterations is almost an alternate mechanism like mutations for the development of cancer Hence we can exploit this mechanism for prevention and therapy Of course mutation is not reversible but epigenetic changes are reversible Since there are several changes we get the option of individual and combined targets for intervention Since many changes occur during the early stages of cancer these are ideal for preventive measures Reversal of gene silencing potentially can alter several signaling pathways making a significant contribution in managing the different stages of cancer

Strategies for cancer prevention and therapy DNA methylation inhibitors 5-azacytidine 5-aza-2 -deoxycytidine (both are demethylating agents). Efficacy of aza-cytidines correlates with the acute reversal of gene silencing. DNA methylation inhibitors (DNMTi) are usually the nucleoside analog family members. Inside the cells they are converted into deoxynucleotide triphosphates and are incorporated into replicating DNA in place of cytosine I have mentioned about these drugs and HDAC inhibitors in another lecture on molecularly targeted drugs for cancer therapy Similarly HDAC inhibitors are also already in the clinic. They generally have antitumor, growth inhibitory, proapoptotic, and prodifferentiation properties They upregulate the expression of p21 (cell cycle inhibitor) due to hyper acetylation of its promoter by these drugs. Suberoylanilide hydroxamic acid (SAHA), also known as vorinostat, has been approved for the treatment of T cell cutaneous lymphoma Other promising hydroxamic acid derivatives are Trichostatin A, Panobinostat and Belinostat Apart from these some short chain fatty acids such as sodium butyrate, phenyl acetate, phenyl butyrate, valproate also act as HDAC inhibitors Others include certain cyclic peptides and benzamide derivatives Many other compounds are being developed as drugs either alone or in combination with other known drugs. Inhibitors of demethylases (HDMs) Pargyline Phenelzine Tranylcypromine Inhibitors of methyltransferases (HMTs) Chaetocin DZNep Allantodapsone

Inhibitors of histone acetyltransferases (HATs) Garcinol Curcumin Anacardic acid Sirtuin (class III HDAC) inhibitors such as sirtinol are gaining importance as well since they induce p53 activity and thus induce apoptosis Biogenesis of mirnas MicroRNAs (mirnas) are noncoding RNAs (ncrnas) of 18-25 nucleotides in length They are fine regulators of gene expression and hence affect several biological processes such as development, differentiation, and cell cycle regulation to senescence and metabolism mirna genes are transcribed like other genes in the nucleus with RNA polymerase II The primary transcript called pri-mirna is very long with the cap and poly A tail etc similar to the protein-coding genes They are further processed by a ribonuclease called Drosha and its partner DGCR8 to form pre-mirnas This is exported from the nucleus in the presence of exportin 5 This is further cleaved by another ribonuclease called Dicer to form a duplex RNA that binds to a complex called RISC containing many proteins including the Argonaute family of proteins One of the strands get separated and acts as mirna while the other strand gets degraded. The mature mirna binds with a complimentary sequence present on the 3 UTR of the target gene mrnas If there is a perfect complimentarity this results in the degradation of the target mrna and if there is partial complimentarity it results in translational inhibition thus silencing the target gene expression in both cases Interestingly some mirnas are found to be oncogenic while many other mirnas act like tumor suppressors

Epigenetic regulation of mirnas in human cancer Epigenetics plays an important role in the aberrant expression of mirnas in many cancers The expression levels of several mirnas change upon treatment with HDAC inhibitors mir-127 is epigenetically silenced by promoter hypermethylation and histone modifications in many cancer cells Acute lymphoblastic leukemia patients have shown a signature expression of 13 mirnas in the CpG islands and high levels of K9H3me2 5-aza-2 - deoxycytidine increased at least one mirna in many ALL patients. mir-124a is methylated in many ALL patients, and its promoter hypermethylation is associated with a higher relapse rate and mortality rate mirnas as regulators of epigenetic effectors mirna 29 targets DNA methyltransferases in lung cancer cell lines. HDAC4 is a direct target of both mir-1 and mir-140. mir-449a binds to the 3 -UTR region of HDAC1. mir-101 directly targets EZH2 (enhancer of zeste homolog 2), a histone methyl transferase, in both prostate and bladder cancers. EZH2 is the catalytic subunit of the polycomb repressive complex 2 (PRC2) and is responsible for heterochromatin formation by trimethylating histone H3 lysine 27 (H3K27me3), leading to the silencing of several TSGs

Study Questions 1. What are the mechanisms by which epigenetic changes affect gene expression? 2. What are mirnas? 3. Low levels of folate in the diet can affect a) Acetylation b) ubiquitination c) methylation d) sumoylation 4. Match the following HAT Waddington mir-1 Methylation S-adenosyl methione HDAC4 epigenetics Curcumin 5. Oxidative deamination of methylated cytosine results in the formation of --------