Sveriges 100 mest populära podcasts

Epigenetics Podcast

Epigenetics Podcast

Discover the stories behind the science!

Prenumerera

iTunes / Overcast / RSS

Webbplats

activemotif.podbean.com

Avsnitt

Comparing CUT&Tag to ENCODE ChIP-Seq in Alzheimer's Disease Samples (Sarah Marzi)

In this episode of the Epigenetics Podcast, we talked with Sarah Marzi from the UK Dementia Research Institute at Imperial College London about her work on epigenetic changes in Alzheimer's Disease, and comparing CUT&Tag to ENCODE ChIP-Seq using limited cell samples.

The interview discusses Sarah Marzi's work on ChIP-Seq experiments and their significance in understanding Alzheimer's disease from an epigenetic perspective. The discussion touches on the widespread dysregulation and changes in acetylation, particularly in genes associated with Alzheimer's risk, providing insights into potential links between epigenetic insults and disease onset.

Moving on to the technical aspects of the study, the interview examines the strategic use of CUT&Tag. It explores the challenges and optimizations involved in accurately profiling limited cell samples. The dialogue also compares CUT&Tag to ENCODE ChIP-Seq, highlighting the complexities of peak calling and data interpretation across different methodologies.

 

References

Kumsta, R., Marzi, S., Viana, J. et al. Severe psychosocial deprivation in early childhood is associated with increased DNA methylation across a region spanning the transcription start site of CYP2E1. Transl Psychiatry 6, e830 (2016). https://doi.org/10.1038/tp.2016.95

Marzi, S. J., Schilder, B. M., Nott, A., Frigerio, C. S., Willaime?Morawek, S., Bucholc, M., Hanger, D. P., James, C., Lewis, P. A., Lourida, I., Noble, W., Rodriguez?Algarra, F., Sharif, J., Tsalenchuk, M., Winchester, L. M., Yaman, Ü., Yao, Z., The Deep Dementia Phenotyping (DEMON) Network, Ranson, J. M., & Llewellyn, D. J. (2023). Artificial intelligence for neurodegenerative experimental models. Alzheimer?s & Dementia, 19(12), 5970?5987. https://doi.org/10.1002/alz.13479

Marzi, S. J., Leung, S. K., Ribarska, T., Hannon, E., Smith, A. R., Pishva, E., Poschmann, J., Moore, K., Troakes, C., Al-Sarraj, S., Beck, S., Newman, S., Lunnon, K., Schalkwyk, L. C., & Mill, J. (2018). A histone acetylome-wide association study of Alzheimer?s disease identifies disease-associated H3K27ac differences in the entorhinal cortex. Nature Neuroscience, 21(11), 1618?1627. https://doi.org/10.1038/s41593-018-0253-7

Hu, D., Abbasova, L., Schilder, B. M., Nott, A., Skene, N. G., & Marzi, S. J. (2022). CUT&Tag recovers up to half of ENCODE ChIP-seq peaks in modifications of H3K27 [Preprint]. Genomics. https://doi.org/10.1101/2022.03.30.486382

 

Related Episodes

When is a Peak a Peak? (Claudio Cantù)

Development of Integrative Machine Learning Tools for Neurodegenerative Diseases (Enrico Glaab)

DNA Methylation Alterations in Neurodegenerative Diseases (Paula Desplats)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on X

Active Motif on LinkedIn

Email: [email protected]

2024-04-18
Länk till avsnitt

The Role of Hat1p in Chromatin Assembly (Mark Parthun)

In this episode of the Epigenetics Podcast, we talked with Mark Parthun from Ohio State University about his work on the role of Hat1p in chromatin assembly.

Mark Parthun shares insights into his pivotal paper in 2004 that explored the link between type B histone acetyltransferases and chromatin assembly, setting the stage for his current research interests in epigenetics. He highlights the role of HAT1 in acetylating lysines on newly synthesized histones, its involvement in double-strand break repair, and the search for phenotypes associated with HAT1 mutations.

The discussion expands to a collaborative research project between two scientists uncovering the roles of HAT1 and NASP as chaperones in chromatin assembly. Transitioning from yeast to mouse models, the team investigated the effects of HAT1 knockout on mouse phenotypes, particularly in lung development and craniofacial morphogenesis. They also explored the impact of histone acetylation on chromatin dynamics and its influence on lifespan, aging processes, and longevity.

 

References

Parthun, M. R., Widom, J., & Gottschling, D. E. (1996). The Major Cytoplasmic Histone Acetyltransferase in Yeast: Links to Chromatin Replication and Histone Metabolism. Cell, 87(1), 85?94. https://doi.org/10.1016/S0092-8674(00)81325-2

Kelly, T. J., Qin, S., Gottschling, D. E., & Parthun, M. R. (2000). Type B histone acetyltransferase Hat1p participates in telomeric silencing. Molecular and cellular biology, 20(19), 7051?7058. https://doi.org/10.1128/MCB.20.19.7051-7058.2000

Ai, X., & Parthun, M. R. (2004). The nuclear Hat1p/Hat2p complex: a molecular link between type B histone acetyltransferases and chromatin assembly. Molecular cell, 14(2), 195?205. https://doi.org/10.1016/s1097-2765(04)00184-4

Nagarajan, P., Ge, Z., Sirbu, B., Doughty, C., Agudelo Garcia, P. A., Schlederer, M., Annunziato, A. T., Cortez, D., Kenner, L., & Parthun, M. R. (2013). Histone acetyl transferase 1 is essential for mammalian development, genome stability, and the processing of newly synthesized histones H3 and H4. PLoS genetics, 9(6), e1003518. https://doi.org/10.1371/journal.pgen.1003518

Agudelo Garcia, P. A., Hoover, M. E., Zhang, P., Nagarajan, P., Freitas, M. A., & Parthun, M. R. (2017). Identification of multiple roles for histone acetyltransferase 1 in replication-coupled chromatin assembly. Nucleic Acids Research, 45(16), 9319?9335. https://doi.org/10.1093/nar/gkx545

Popova, L. V., Nagarajan, P., Lovejoy, C. M., Sunkel, B. D., Gardner, M. L., Wang, M., Freitas, M. A., Stanton, B. Z., & Parthun, M. R. (2021). Epigenetic regulation of nuclear lamina-associated heterochromatin by HAT1 and the acetylation of newly synthesized histones. Nucleic Acids Research, 49(21), 12136?12151. https://doi.org/10.1093/nar/gkab1044

 

Related Episodes

Regulation of Chromatin Organization by Histone Chaperones (Geneviève Almouzni)

Effects of Non-Enzymatic Covalent Histone Modifications on Chromatin (Yael David)

scDamID, EpiDamID and Lamina Associated Domains (Jop Kind)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on X

Active Motif on LinkedIn

Email: [email protected]

2024-04-04
Länk till avsnitt

The Impact of Paternal Diet on Offspring Metabolism (Upasna Sharma)

In this episode of the Epigenetics Podcast, we talked with Upasna Sharma from UC Santa Cruz about her work on a number of interesting projects on H2A.Z and telomeres, the impact of paternal diet on offspring metabolism, and the role of small RNAs in sperm.

In this interview Upasna Sharma discusses her work on the study of the paternal diet's impact on offspring metabolism. She reveals the discovery of small non-coding RNAs, particularly tRNA fragments, in mature mammalian sperm that may carry epigenetic information to the next generation. She explains the specific alterations in tRNA fragment levels in response to a low-protein diet and the connections found between tRNA fragments and metabolic status.

Dr. Sharma further explains the degradation and stabilization of tRNA fragments in cells and the processes involved in their regulation. She shares their observation of tRNA fragment abundance in epididymal sperm, despite the sperm being transcriptionally silent at that time. This leads to a discussion on the role of the epididymis in the reprogramming of small RNA profiles and the transportation of tRNA fragments through extracellular vesicles.

The conversation then shifts towards the potential mechanism of how environmental information could be transmitted to sperm and the observed changes in small RNAs in response to a low-protein diet. Dr. Sharma discusses the manipulation of small RNAs in embryos and mouse embryonic stem cells, revealing their role in regulating specific sets of genes during early development. However, the exact mechanisms that link these early changes to metabolic phenotypes are still being explored.

References

Sharma, U., Conine, C. C., Shea, J. M., Boskovic, A., Derr, A. G., Bing, X. Y., Belleannee, C., Kucukural, A., Serra, R. W., Sun, F., Song, L., Carone, B. R., Ricci, E. P., Li, X. Z., Fauquier, L., Moore, M. J., Sullivan, R., Mello, C. C., Garber, M., & Rando, O. J. (2016). Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science (New York, N.Y.), 351(6271), 391?396. https://doi.org/10.1126/science.aad6780

Sharma, U., Sun, F., Conine, C. C., Reichholf, B., Kukreja, S., Herzog, V. A., Ameres, S. L., & Rando, O. J. (2018). Small RNAs Are Trafficked from the Epididymis to Developing Mammalian Sperm. Developmental cell, 46(4), 481?494.e6. https://doi.org/10.1016/j.devcel.2018.06.023

Rinaldi, V. D., Donnard, E., Gellatly, K., Rasmussen, M., Kucukural, A., Yukselen, O., Garber, M., Sharma, U., & Rando, O. J. (2020). An atlas of cell types in the mouse epididymis and vas deferens. eLife, 9, e55474. https://doi.org/10.7554/eLife.55474

 

Related Episodes

The Epigenetics of Human Sperm Cells (Sarah Kimmins)

Transgenerational Inheritance and Evolution of Epimutations (Peter Sarkies)

The Role of Small RNAs in Transgenerational Inheritance in C. elegans (Oded Rechavi)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on X

Active Motif on LinkedIn

Email: [email protected]

2024-03-21
Länk till avsnitt

H3K36me3, H4K16ac and Cryptic Transcription in Ageing (Weiwei Dang)

In this episode of the Epigenetics Podcast, we talked with Weiwei Dang from Baylor College of Medicine about his work on molecular mechanisms of aging and the role of H3K36me3 and cryptic transcription in cellular aging.

The team in the Weiwei Dang lab explored the connection between histone marks, specifically H4K16 acetylation and H3K36 methylation, and aging. Dr. Dang describes how the lab conducted experiments by mutating H4K16 to determine its effect on lifespan. They observed that the mutation to glutamine accelerated the aging process and shortened lifespan, providing causal evidence for the relationship between H4K16 and lifespan. They also discovered that mutations in acetyltransferase and demethylase enzymes had opposite effects on lifespan, further supporting a causal relationship.

Weiwei Dang then discusses their expanded research on aging, conducting high-throughput screens to identify other histone residues and mutants in yeast that regulate aging. They found that most mutations at K36 shortened lifespan, and so they decided to follow up on a site that is known to be methylated and play a role in gene function. They discovered that H3K36 methylation helps suppress cryptic transcription, which is transcription that initiates from within the gene rather than at the promoter. Mutants lacking K36 methylation showed an aging phenotype. They also found evidence of cryptic transcription in various datasets related to aging and senescence, including C. elegans and mammalian cells.

References

Dang, W., Steffen, K., Perry, R. et al. Histone H4 lysine 16 acetylation regulates cellular lifespan. Nature 459, 802?807 (2009). https://doi.org/10.1038/nature08085

Sen, P., Dang, W., Donahue, G., Dai, J., Dorsey, J., Cao, X., Liu, W., Cao, K., Perry, R., Lee, J. Y., Wasko, B. M., Carr, D. T., He, C., Robison, B., Wagner, J., Gregory, B. D., Kaeberlein, M., Kennedy, B. K., Boeke, J. D., & Berger, S. L. (2015). H3K36 methylation promotes longevity by enhancing transcriptional fidelity. Genes & development, 29(13), 1362?1376. https://doi.org/10.1101/gad.263707.115

Yu, R., Cao, X., Sun, L. et al. Inactivating histone deacetylase HDA promotes longevity by mobilizing trehalose metabolism. Nat Commun 12, 1981 (2021). https://doi.org/10.1038/s41467-021-22257-2

McCauley, B.S., Sun, L., Yu, R. et al. Altered chromatin states drive cryptic transcription in aging mammalian stem cells. Nat Aging 1, 684?697 (2021). https://doi.org/10.1038/s43587-021-00091-x

 

Related Episodes

Epigenetic Mechanisms of Aging and Longevity (Shelley Berger)

Epigenetic Clocks and Biomarkers of Ageing (Morgan Levine)

Gene Dosage Alterations in Evolution and Ageing (Claudia Keller Valsecchi)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on X

Active Motif on LinkedIn

Email: [email protected]

2024-03-07
Länk till avsnitt

Split-Pool Recognition of Interactions by Tag Extension (SPRITE) (Mitch Guttman)

In this episode of the Epigenetics Podcast, we talked with Mitch Guttman from California Institute of Technology about his work on characterising the 3D interactions of the genome using Split-Pool Recognition of Interactions by Tag Extension (SPRITE).

Mitch Guttman discusses his exploration of the long non-coding RNA Xist, which plays a crucial role in X chromosome inactivation. He explains how they discovered that Xist is present everywhere in the nucleus, not just in specific locations on the X chromosome. Through their research, they identified critical proteins like SHARP that are involved in X chromosome silencing.

The discussion then shifts to SPRITE, a method they developed to map multi-way contacts and generalize beyond DNA to include RNA and proteins. They compare SPRITE to classical proximity ligation methods like Hi-C and discuss how cluster sizes in SPRITE can estimate 3D distances between molecules. The conversation also touches upon the potential of applying SPRITE to single-cell experiments, allowing for the mapping of higher order nucleic acid interactions and tracking the connectivity of DNA fragments in individual cells.

 

References

Jesse M. Engreitz et al., The Xist lncRNA Exploits Three-Dimensional Genome Architecture to Spread Across the X Chromosome. Science 341,1237973(2013). DOI:10.1126/science.1237973

Chun-Kan Chen et al., Xist recruits the X chromosome to the nuclear lamina to enable chromosome-wide silencing. Science 354, 468-472(2016). DOI: 10.1126/science.aae0047

Quinodoz, S. A., Ollikainen, N., Tabak, B., Palla, A., Schmidt, J. M., Detmar, E., Lai, M. M., Shishkin, A. A., Bhat, P., Takei, Y., Trinh, V., Aznauryan, E., Russell, P., Cheng, C., Jovanovic, M., Chow, A., Cai, L., McDonel, P., Garber, M., & Guttman, M. (2018). Higher-Order Inter-chromosomal Hubs Shape 3D Genome Organization in the Nucleus. Cell, 174(3), 744-757.e24. https://doi.org/10.1016/j.cell.2018.05.024

Goronzy, I. N., Quinodoz, S. A., Jachowicz, J. W., Ollikainen, N., Bhat, P., & Guttman, M. (2022). Simultaneous mapping of 3D structure and nascent RNAs argues against nuclear compartments that preclude transcription. Cell Reports, 41(9), 111730. https://doi.org/10.1016/j.celrep.2022.111730

Perez, A. A., Goronzy, I. N., Blanco, M. R., Guo, J. K., & Guttman, M. (2023). ChIP-DIP: A multiplexed method for mapping hundreds of proteins to DNA uncovers diverse regulatory elements controlling gene expression [Preprint]. Genomics. https://doi.org/10.1101/2023.12.14.571730

 

Related Episodes

Epigenetics and X-Inactivation (Edith Heard)

Hi-C and Three-Dimensional Genome Sequencing (Erez Lieberman Aiden)

Unraveling Mechanisms of Chromosome Formation (Job Dekker)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on X

Active Motif on LinkedIn

Email: [email protected]

2024-02-22
Länk till avsnitt

MLL Proteins in Mixed-Lineage Leukemia (Yali Dou)

In this episode of the Epigenetics Podcast, we talked with Yali Dou from Keck School of Medicine of USC about her work on MLL Proteins in Mixed-Lineage Leukemia.

To start off this Interview Yali describes her early work on MLL1 and its function in transcription, particularly its involvement in histone modification. She explains her successful purification of the MLL complex and the discovery of MOF as one of the proteins involved.

Next, the interview focuses on her work in reconstituting the MLL core complex and the insights gained from this process. She shares her experience of reconstituting the MLL complex and discusses her focus on the crosstalk of H3K4 and H3K79 methylation, regulated by H2BK34 ubiquitination.

The podcast then delves into the therapeutic potential of MLL1, leading to the discovery of a small molecule inhibitor. Finally, we talk about the importance of the protein WDR5 in the assembly of MLL complexes and how targeting the WDR5-ML interaction can inhibit MLL activity.

 

References

Dou, Y., Milne, T., Ruthenburg, A. et al. Regulation of MLL1 H3K4 methyltransferase activity by its core components. Nat Struct Mol Biol 13, 713?719 (2006). https://doi.org/10.1038/nsmb1128

Wu, L., Zee, B. M., Wang, Y., Garcia, B. A., & Dou, Y. (2011). The RING Finger Protein MSL2 in the MOF Complex Is an E3 Ubiquitin Ligase for H2B K34 and Is Involved in Crosstalk with H3 K4 and K79 Methylation. Molecular Cell, 43(1), 132?144. https://doi.org/10.1016/j.molcel.2011.05.015

Cao, F., Townsend, E. C., Karatas, H., Xu, J., Li, L., Lee, S., Liu, L., Chen, Y., Ouillette, P., Zhu, J., Hess, J. L., Atadja, P., Lei, M., Qin, Z. S., Malek, S., Wang, S., & Dou, Y. (2014). Targeting MLL1 H3K4 Methyltransferase Activity in Mixed-Lineage Leukemia. Molecular Cell, 53(2), 247?261. https://doi.org/10.1016/j.molcel.2013.12.001

Park, S.H., Ayoub, A., Lee, YT. et al. Cryo-EM structure of the human MLL1 core complex bound to the nucleosome. Nat Commun 10, 5540 (2019). https://doi.org/10.1038/s41467-019-13550-2

 

Related Episodes

Dosage Compensation in Drosophila (Asifa Akhtar)

Targeting COMPASS to Cure Childhood Leukemia (Ali Shilatifard)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on X

Active Motif on LinkedIn

Email: [email protected]

2024-02-08
Länk till avsnitt

Sex-biased Imprinting and DNA Regulatory Landscapes During Reprogramming (Sam Buckberry)

In this episode of the Epigenetics Podcast, we talked with Sam Buckberry from the Telethon Kids Institute about his work on gene imprinting, sex-biased gene expression, DNA regulatory landscapes, and genomics in the indigenous population of Australia.

Sam Buckberry's research career started with working on the imprinting of H19, IGF2, and IGF2R genes in the placenta. We talk about the controversy surrounding the imprinting of IGF2R and how his study used pyrosequencing to quantify gene expression. We also discuss Sam's work on sex-biased gene expression in the placenta and the identification of a cluster of genes related to placental development and pregnancy.

In addition, we talk about Sam's research on reprogramming and the characterization of DNA regulatory landscapes during the process. We discuss the challenges of working with sequencing data, the discovery of epigenetic memories, and erasing them during reprogramming. Towards the end of the conversation, Sam mentions his current work in setting up an epigenetics group focused on indigenous genomics. They are conducting a large-scale, multi-omics study on cardiometabolic conditions in samples from indigenous Australian communities, with the goal of identifying biomarkers and better understanding the molecular basis of these conditions.

 

References

Buckberry, S., Liu, X., Poppe, D. et al. Transient naive reprogramming corrects hiPS cells functionally and epigenetically. Nature 620, 863?872 (2023). https://doi.org/10.1038/s41586-023-06424-7

Knaupp AS1, Buckberry S1, Pflueger J, Lim SM, Ford E, Larcombe MR, Rossello FJ, de Mendoza A, Alaei S, Firas J, Holmes ML, Nair SS, Clark SJ, Nefzger CM, Lister R and Polo JM (2017). Transient and permanent reconfiguration of chromatin and transcription factor occupancy drive reprogramming. Cell Stem Cell 21, 1-12 1 Co-first author

 

Related Episodes

The Effect of Mechanotransduction on Chromatin Structure and Transcription in Stem Cells (Sara Wickström)

Differential Methylated Regions in Autism Spectrum Disorders (Janine La Salle)

The Role of Pioneer Factors Zelda and Grainyhead at the Maternal-to-Zygotic Transition (Melissa Harrison)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on X

Active Motif on LinkedIn

Email: [email protected]

2024-01-25
Länk till avsnitt

BET Proteins and Their Role in Chromosome Folding and Compartmentalization (Kyle Eagen)

In this episode of the Epigenetics Podcast, we talked with Kyle Eagen from Baylor College of Medicine about his work on BET Proteins and their role in chromosome folding and compartmentalization.

In the early days of his research career Dr. Eagen made use of genomics and microscopy to study chromosomes, particularly polytene chromosomes in Drosophila. The correlation between the folding patterns detected by Hi-C and polytene bands highlights the similarities between the two, bridging traditional cytology with modern NGS methods. This work formed the basis of Kyle's thesis and sparked his interest in nuclear organization and chromosome 3D structure.

In his independent lab Kyle then studied compartments in chromatin structure and focused on the relationship between histone modifications and the 3D structure of chromosomes. The discovery of BRD4-NUT, a fusion oncoprotein that reprograms chromosome 3D structure, is highlighted as a significant step forward in understanding chromatin structure.

The conversation then shifts to the use of a tool to test hypotheses about the involvement of BRD4 in a specific process, leading to consistent results and considerations for manipulating chromosome organization for therapeutic purposes. The role of BET proteins in genome folding and the need for further research on other factors involved in 3D genome structure are discussed.

 

References

Rosencrance, C. D., Ammouri, H. N., Yu, Q., Ge, T., Rendleman, E. J., Marshall, S. A., & Eagen, K. P. (2020). Chromatin Hyperacetylation Impacts Chromosome Folding by Forming a Nuclear Subcompartment. Molecular Cell, 78(1), 112-126.e12. https://doi.org/10.1016/j.molcel.2020.03.018

Huang, Y., Durall, R. T., Luong, N. M., Hertzler, H. J., Huang, J., Gokhale, P. C., Leeper, B. A., Persky, N. S., Root, D. E., Anekal, P. V., Montero Llopis, P. D. L. M., David, C. N., Kutok, J. L., Raimondi, A., Saluja, K., Luo, J., Zahnow, C. A., Adane, B., Stegmaier, K., ? French, C. A. (2023). EZH2 Cooperates with BRD4-NUT to Drive NUT Carcinoma Growth by Silencing Key Tumor Suppressor Genes. Cancer Research, 83(23), 3956?3973. https://doi.org/10.1158/0008-5472.CAN-23-1475

 

Related Episodes

Hi-C and Three-Dimensional Genome Sequencing (Erez Lieberman Aiden)

Genome Organization Mediated by RNA Polymerase II (Argyrys Papantonis)

Analysis of 3D Chromatin Structure Using Super-Resolution Imaging (Alistair Boettiger)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on X

Active Motif on LinkedIn

Email: [email protected]

2024-01-11
Länk till avsnitt

Epigenetic Underpinnings of Human Addiction (Francesca Telese & Jessica Zhou)

In this episode of the Epigenetics Podcast, we talked with Francesca Telese from UC San Diego and Jessica Zhou from Cold Spring Harbour about their work on the molecular underpinnings of human addiction.

Francesca Telese worked on neuronal enhancers and their pivotal role in governing gene activity. She sheds light on her remarkable findings concerning the epigenetic signature of neuronal enhancers that are intricately involved in synaptic plasticity.

Jessica Zhou joined Francesca Telese's lab as a PhD student where she worked on elucidating the effects of chronic cannabis use on memory and behavior in mice. She takes us through the fascinating correlation between THC and gene co-expression networks. Francesca and Jessicathen discuss the utilization of genetically diverse outbred rats in their research, along with the crucial exploration of cell type specificity in gene expression studies. They then delve into the long-term changes that occur in the brain after drug exposure and the profound implications for relapse. Additionally, they touch upon the challenges they face in analyzing single-cell data.

 

References

Zhou, J. L., de Guglielmo, G., Ho, A. J., Kallupi, M., Pokhrel, N., Li, H. R., Chitre, A. S., Munro, D., Mohammadi, P., Carrette, L. L. G., George, O., Palmer, A. A., McVicker, G., & Telese, F. (2023). Single-nucleus genomics in outbred rats with divergent cocaine addiction-like behaviors reveals changes in amygdala GABAergic inhibition. Nature neuroscience, https://doi.org/10.1038/s41593-023-01452-y

Wang, J., Telese, F., Tan, Y., Li, W., Jin, C., He, X., Basnet, H., Ma, Q., Merkurjev, D., Zhu, X., Liu, Z., Zhang, J., Ohgi, K., Taylor, H., White, R. R., Tazearslan, C., Suh, Y., Macfarlan, T. S., Pfaff, S. L., & Rosenfeld, M. G. (2015). LSD1n is an H4K20 demethylase regulating memory formation via transcriptional elongation control. Nature neuroscience, 18(9), 1256?1264. https://doi.org/10.1038/nn.4069

 

Related Episodes

The Role of Histone Dopaminylation and Serotinylation in Neuronal Plasticity (Ian Maze)

 

Contact

Epigenetics Podcast on X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Epigenetics Podcast on Threads

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-12-21
Länk till avsnitt

H3K79 Methylation, DOT1L, and FOXG1 in Neural Development (Tanja Vogel)

In this episode of the Epigenetics Podcast, we talked with Tanja Vogel from the University Clinics Freiburg about her work on epigenetic modifications in stem cells during central nervous system development.

During our discussion, Dr. Vogel shared that she and her team have investigated H3K79 methylation and its functional significance, which remains a topic of debate in the scientific community. They?ve also investigated the role of DOT1L in neural development and its implications for neuronal networks, as disrupting DOT1L can lead to conditions such as epilepsy and schizophrenia. They explored the function of the SOX2 enhancer in the presence or absence of DOT1L enzymatic inhibition.

The conversation then shifts to FoxG1, a vital player in forebrain development. The team uncovered its role in chromatin accessibility and its connection to microRNA processing. Their study, utilizing ChIP-Seq, reveals FoxG1's interactions with enhancer regions and other transcription factors, like NeuroD1.

 

### References

Britanova, O., de Juan Romero, C., Cheung, A., Kwan, K. Y., Schwark, M., Gyorgy, A., Vogel, T., Akopov, S., Mitkovski, M., Agoston, D., Sestan, N., Molnár, Z., & Tarabykin, V. (2008). Satb2 is a postmitotic determinant for upper-layer neuron specification in the neocortex. Neuron, 57(3), 378?392. https://doi.org/10.1016/j.neuron.2007.12.028

Büttner, N., Johnsen, S. A., Kügler, S., & Vogel, T. (2010). Af9/Mllt3 interferes with Tbr1 expression through epigenetic modification of histone H3K79 during development of the cerebral cortex. Proceedings of the National Academy of Sciences of the United States of America, 107(15), 7042?7047. https://doi.org/10.1073/pnas.0912041107

Franz, H., Villarreal, A., Heidrich, S., Videm, P., Kilpert, F., Mestres, I., Calegari, F., Backofen, R., Manke, T., & Vogel, T. (2019). DOT1L promotes progenitor proliferation and primes neuronal layer identity in the developing cerebral cortex. Nucleic acids research, 47(1), 168?183. https://doi.org/10.1093/nar/gky953

Ferrari, F., Arrigoni, L., Franz, H., Izzo, A., Butenko, L., Trompouki, E., Vogel, T., & Manke, T. (2020). DOT1L-mediated murine neuronal differentiation associates with H3K79me2 accumulation and preserves SOX2-enhancer accessibility. Nature communications, 11(1), 5200. https://doi.org/10.1038/s41467-020-19001-7

Akol, I., Izzo, A., Gather, F., Strack, S., Heidrich, S., Ó hAilín, D., Villarreal, A., Hacker, C., Rauleac, T., Bella, C., Fischer, A., Manke, T., & Vogel, T. (2023). Multimodal epigenetic changes and altered NEUROD1 chromatin binding in the mouse hippocampus underlie FOXG1 syndrome. Proceedings of the National Academy of Sciences of the United States of America, 120(2), e2122467120. https://doi.org/10.1073/pnas.2122467120

 

Related Episodes

Molecular Mechanisms of Chromatin Modifying Enzymes (Karim-Jean Armache)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-11-30
Länk till avsnitt

Function of Insulators in 3D Genome Folding (Maria Gambetta)

In this episode of the Epigenetics Podcast, we talked with Maria Gambetta from the University of Lausanne about her work on the function of insulators in 3D genome folding.

Maria Gambetta focuses on investigating 3D contact dynamics between enhancers and promoters, providing insights into tissue-specific gene activation. The team used capture-C to analyze dynamic looping events, emphasizing the significance of accessible chromatin peaks in enhancer-promoter interactions. Furthermore, they focused on gene insulation and CTCF's role in forming topologically associating domains in Drosophila. Hi-C analysis on CTCF mutants revealed the conservation of TAD boundary mechanisms, identifying CP-190 as a potential binding protein.

Their findings on the loss of TAD boundaries in mutants and the role of transcription in TAD boundary formation are discussed as well as the function of CP190 and insulators in preventing interactions between promoters and enhancers. Their work challenges existing models of insulator function and seeks to understand their mechanisms better. The conversation concludes with insights into long-range regulatory associations in Drosophila, emphasizing the punctual interactions between transcription factor binding sites and their effect on neural gene transcription and genome folding.

 

References

Gambetta, M. C., Oktaba, K., & Müller, J. (2009). Essential role of the glycosyltransferase sxc/Ogt in polycomb repression. Science (New York, N.Y.), 325(5936), 93?96. https://doi.org/10.1126/science.1169727

Kaushal, A., Mohana, G., Dorier, J., Özdemir, I., Omer, A., Cousin, P., Semenova, A., Taschner, M., Dergai, O., Marzetta, F., Iseli, C., Eliaz, Y., Weisz, D., Shamim, M. S., Guex, N., Lieberman Aiden, E., & Gambetta, M. C. (2021). CTCF loss has limited effects on global genome architecture in Drosophila despite critical regulatory functions. Nature communications, 12(1), 1011. https://doi.org/10.1038/s41467-021-21366-2

Hoencamp, C., Dudchenko, O., Elbatsh, A. M. O., Brahmachari, S., Raaijmakers, J. A., van Schaik, T., Sedeño Cacciatore, Á., Contessoto, V. G., van Heesbeen, R. G. H. P., van den Broek, B., Mhaskar, A. N., Teunissen, H., St Hilaire, B. G., Weisz, D., Omer, A. D., Pham, M., Colaric, Z., Yang, Z., Rao, S. S. P., Mitra, N., ? Rowland, B. D. (2021). 3D genomics across the tree of life reveals condensin II as a determinant of architecture type. Science (New York, N.Y.), 372(6545), 984?989. https://doi.org/10.1126/science.abe2218

Mohana, G., Dorier, J., Li, X., Mouginot, M., Smith, R. C., Malek, H., Leleu, M., Rodriguez, D., Khadka, J., Rosa, P., Cousin, P., Iseli, C., Restrepo, S., Guex, N., McCabe, B. D., Jankowski, A., Levine, M. S., & Gambetta, M. C. (2023). Chromosome-level organization of the regulatory genome in the Drosophila nervous system. Cell, 186(18), 3826?3844.e26. https://doi.org/10.1016/j.cell.2023.07.008

 

Related Episodes

Hi-C and Three-Dimensional Genome Sequencing (Erez Lieberman Aiden)

Biophysical Modeling of 3-D Genome Organization (Leonid Mirny)

Long-Range Transcriptional Control by 3D Chromosome Structure (Luca Giorgetti)

 

Contact

Epigenetics Podcast on Twitter/X

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-11-16
Länk till avsnitt

Contribution of the Estrogen Receptor to Breast Cancer Progression (Jason Carroll)

In this episode of the Epigenetics Podcast, we talked with Jason Carroll from the Cambridge Research Institute about his work on contribution of estrogen receptor to breast cancer progression.

The Podcast centers around the crucial role of the forkhead protein FOXA1 in breast cancer. FOXA1 acts as a pioneer transcription factor, facilitating gene regulation by recruiting nuclear receptors to chromatin, profoundly influencing gene expression in various breast cancer subtypes. The FOXA1-positive subtype of triple-negative breast cancer, despite being estrogen receptor-negative, shares gene expression profiles with estrogen receptor-positive breast cancer, shedding light on the importance of targeting the androgen receptor for treatment.

The challenges of studying transcription factor mappings from clinical samples are explored, with a focus on the ChIP-seq method's success in mapping estrogen receptor binding sites. Various techniques for transcription factor mapping, including CUT&RUN, CUT&Tag, and ChIP-exo, are discussed, as well as the potential of mass spec techniques like the RIME method in analyzing protein interactions. An intriguing experiment involving the purification of multiple proteins to identify interactions is highlighted.

 

References

Carroll, J. S., Meyer, C. A., Song, J., Li, W., Geistlinger, T. R., Eeckhoute, J., Brodsky, A. S., Keeton, E. K., Fertuck, K. C., Hall, G. F., Wang, Q., Bekiranov, S., Sementchenko, V., Fox, E. A., Silver, P. A., Gingeras, T. R., Liu, X. S., & Brown, M. (2006). Genome-wide analysis of estrogen receptor binding sites. Nature genetics, 38(11), 1289?1297. https://doi.org/10.1038/ng1901

Hurtado, A., Holmes, K. A., Geistlinger, T. R., Hutcheson, I. R., Nicholson, R. I., Brown, M., Jiang, J., Howat, W. J., Ali, S., & Carroll, J. S. (2008). Regulation of ERBB2 by oestrogen receptor-PAX2 determines response to tamoxifen. Nature, 456(7222), 663?666. https://doi.org/10.1038/nature07483

Ross-Innes, C. S., Stark, R., Teschendorff, A. E., Holmes, K. A., Ali, H. R., Dunning, M. J., Brown, G. D., Gojis, O., Ellis, I. O., Green, A. R., Ali, S., Chin, S. F., Palmieri, C., Caldas, C., & Carroll, J. S. (2012). Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature, 481(7381), 389?393. https://doi.org/10.1038/nature10730

Mohammed, H., Russell, I. A., Stark, R., Rueda, O. M., Hickey, T. E., Tarulli, G. A., Serandour, A. A., Birrell, S. N., Bruna, A., Saadi, A., Menon, S., Hadfield, J., Pugh, M., Raj, G. V., Brown, G. D., D'Santos, C., Robinson, J. L., Silva, G., Launchbury, R., Perou, C. M., ? Carroll, J. S. (2015). Progesterone receptor modulates ER? action in breast cancer. Nature, 523(7560), 313?317. https://doi.org/10.1038/nature14583

 

Related Episodes

Pioneer Transcription Factors and Their Influence on Chromatin Structure (Ken Zaret)

The Role of Pioneer Factors Zelda and Grainyhead at the Maternal-to-Zygotic Transition (Melissa Harrison)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Epigenetics Podcast on Bluesky

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-11-02
Länk till avsnitt

Inheritance of Transcriptional Memory by Mitotic Bookmarking (Sheila Teves)

In this episode of the Epigenetics Podcast, we caught up with Sheila Teves from the University of British Columbia to talk about her work on the inheritance of transcriptional memory by mitotic bookmarking.

Early in her research career, Sheila Teves focused on the impact of nucleosomes on torsional stress and gene regulation. She also highlights the development of a genome-wide approach to measure torsional stress and its relationship to nucleosome dynamics and RNA polymerase regulation.

The conversation then shifts to her focus on transcriptional memory and mitotic bookmarking during her postdoc in the Tijan lab. She explores the concept of mitotic bookmarking, whereby certain transcription factors remain bound to their target sites during mitosis, facilitating efficient reactivation of transcription after cell division. She discusses her findings on the behavior of transcription factors on mitotic chromosomes, challenging the notion that they are excluded during mitosis. She also discusses the differences in binding behavior between the general transcription factor TBP and other transcription factors. Finally, the effect of formaldehyde fixation on the potential to find transcription factors bound to mitotic chromosomes is discussed.

 

References

Teves, S., Henikoff, S. Transcription-generated torsional stress destabilizes nucleosomes. Nat Struct Mol Biol 21, 88?94 (2014). https://doi.org/10.1038/nsmb.2723

Sheila S Teves, Luye An, Anders S Hansen, Liangqi Xie, Xavier Darzacq, Robert Tjian (2016) A dynamic mode of mitotic bookmarking by transcription factors eLife 5:e22280. https://doi.org/10.7554/eLife.22280

Sheila S Teves, Luye An, Aarohi Bhargava-Shah, Liangqi Xie, Xavier Darzacq, Robert Tjian (2018) A stable mode of bookmarking by TBP recruits RNA polymerase II to mitotic chromosomes eLife 7:e35621. https://doi.org/10.7554/eLife.35621

Kwan, J. Z. J., Nguyen, T. F., Uzozie, A. C., Budzynski, M. A., Cui, J., Lee, J. M. C., Van Petegem, F., Lange, P. F., & Teves, S. S. (2023). RNA Polymerase II transcription independent of TBP in murine embryonic stem cells. eLife, 12, e83810. https://doi.org/10.7554/eLife.83810

Price, R. M., Budzy?ski, M. A., Shen, J., Mitchell, J. E., Kwan, J. Z. J., & Teves, S. S. (2023). Heat shock transcription factors demonstrate a distinct mode of interaction with mitotic chromosomes. Nucleic acids research, 51(10), 5040?5055. https://doi.org/10.1093/nar/gkad304

 

Related Episodes

In Vivo Nucleosome Structure and Dynamics (Srinivas Ramachandran)

From Nucleosome Structure to Function (Karolin Luger)

Structural Analysis of Nucleosomes During Transcription (Lucas Farnung)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-10-19
Länk till avsnitt

Differential Methylated Regions in Autism Spectrum Disorders (Janine La Salle)

In this episode of the Epigenetics Podcast, we talked with Janine La Salle from UC Davis about her work on differential methylated regions in autism spectrum disorders.

In our discussion, Janine LaSalle highlights her work on the placental epigenetic signature, which offers insights into the impact of fetal exposures and gene-environment interactions during the perinatal period. She emphasizes the placenta's value as a surrogate tissue for understanding human diseases. Her research on DNA methylation in the placenta across different mammalian species reveals consistent patterns in partially methylated and highly methylated domains. She explains the critical role of higher methylation levels in specific regions for gene expression and how this knowledge helps trace the placenta's developmental history.

The conversation then delves into Dr. LaSalle's research on the link between placental DNA methylation and autism. Through epigenome-wide association studies, she discovered a novel autism gene and explored the effects of prenatal exposures on DNA methylation profiles. Additionally, she discusses the impact of maternal obesity on offspring neurodevelopment. Ultimately, the goal of her research is to contribute to precision public health and preventative healthcare with epigenetic signatures offering high potential for predicting and preventing future health problems.

 

References

Schroeder, D. I., Blair, J. D., Lott, P., Yu, H. O., Hong, D., Crary, F., Ashwood, P., Walker, C., Korf, I., Robinson, W. P., & LaSalle, J. M. (2013). The human placenta methylome. Proceedings of the National Academy of Sciences of the United States of America, 110(15), 6037?6042. https://doi.org/10.1073/pnas.1215145110

Zhu, Y., Gomez, J. A., Laufer, B. I., Mordaunt, C. E., Mouat, J. S., Soto, D. C., Dennis, M. Y., Benke, K. S., Bakulski, K. M., Dou, J., Marathe, R., Jianu, J. M., Williams, L. A., Gutierrez Fugón, O. J., Walker, C. K., Ozonoff, S., Daniels, J., Grosvenor, L. P., Volk, H. E., Feinberg, J. I., ? LaSalle, J. M. (2022). Placental methylome reveals a 22q13.33 brain regulatory gene locus associated with autism. Genome biology, 23(1), 46. https://doi.org/10.1186/s13059-022-02613-1

Laufer, B. I., Hasegawa, Y., Zhang, Z., Hogrefe, C. E., Del Rosso, L. A., Haapanen, L., Hwang, H., Bauman, M. D., Van de Water, J., Taha, A. Y., Slupsky, C. M., Golub, M. S., Capitanio, J. P., VandeVoort, C. A., Walker, C. K., & LaSalle, J. M. (2022). Multi-omic brain and behavioral correlates of cell-free fetal DNA methylation in macaque maternal obesity models. Nature communications, 13(1), 5538. https://doi.org/10.1038/s41467-022-33162-7

Coulson, R. L., Yasui, D. H., Dunaway, K. W., Laufer, B. I., Vogel Ciernia, A., Zhu, Y., Mordaunt, C. E., Totah, T. S., & LaSalle, J. M. (2018). Snord116-dependent diurnal rhythm of DNA methylation in mouse cortex. Nature communications, 9(1), 1616. https://doi.org/10.1038/s41467-018-03676-0

Neier, K., Grant, T. E., Palmer, R. L., Chappell, D., Hakam, S. M., Yasui, K. M., Rolston, M., Settles, M. L., Hunter, S. S., Madany, A., Ashwood, P., Durbin-Johnson, B., LaSalle, J. M., & Yasui, D. H. (2021). Sex disparate gut microbiome and metabolome perturbations precede disease progression in a mouse model of Rett syndrome. Communications biology, 4(1), 1408. https://doi.org/10.1038/s42003-021-02915-3

 

Related Episodes

DNA Methylation Alterations in Neurodegenerative Diseases (Paula Desplats)

Characterization of Epigenetic States in the Oligodendrocyte Lineage (Gonçalo Castelo-Branco)

The Role of Histone Dopaminylation and Serotinylation in Neuronal Plasticity (Ian Maze)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-10-05
Länk till avsnitt

DNA Damage in Longevity and Ageing (Björn Schumacher)

In this episode of the Epigenetics Podcast, we caught up with Björn Schumacher from the Institute for Genome Stability in Ageing and Disease at the University of Cologne to talk about his work on DNA damage in longevity and ageing.

In this episode Björn Schumacher discusses his research on DNA repair and its impact on ageing. We explore his insights on the effects of DNA damage on transcription, the importance of studying development, and the role of histone modifications. We also discuss paternal DNA damage inheritance and the DREAM complex as a master regulator of DNA repair. The lab?s goal is to enhance somatic DNA repair for healthier ageing and disease prevention.

 

References

Schumacher, B., van der Pluijm, I., Moorhouse, M. J., Kosteas, T., Robinson, A. R., Suh, Y., Breit, T. M., van Steeg, H., Niedernhofer, L. J., van Ijcken, W., Bartke, A., Spindler, S. R., Hoeijmakers, J. H., van der Horst, G. T., & Garinis, G. A. (2008). Delayed and accelerated aging share common longevity assurance mechanisms. PLoS genetics, 4(8), e1000161. https://doi.org/10.1371/journal.pgen.1000161

Ermolaeva, M. A., Segref, A., Dakhovnik, A., Ou, H. L., Schneider, J. I., Utermöhlen, O., Hoppe, T., & Schumacher, B. (2013). DNA damage in germ cells induces an innate immune response that triggers systemic stress resistance. Nature, 501(7467), 416?420. https://doi.org/10.1038/nature12452

Wang, S., Meyer, D. H., & Schumacher, B. (2023). Inheritance of paternal DNA damage by histone-mediated repair restriction. Nature, 613(7943), 365?374. https://doi.org/10.1038/s41586-022-05544-w

Bujarrabal-Dueso, A., Sendtner, G., Meyer, D. H., Chatzinikolaou, G., Stratigi, K., Garinis, G. A., & Schumacher, B. (2023). The DREAM complex functions as conserved master regulator of somatic DNA-repair capacities. Nature structural & molecular biology, 30(4), 475?488. https://doi.org/10.1038/s41594-023-00942-8

 

Related Episodes

Effects of Environmental Cues on the Epigenome and Longevity (Paul Shiels)

Transposable Elements in Gene Regulation and Evolution (Marco Trizzino)

Epigenetic Clocks and Biomarkers of Ageing (Morgan Levine)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-09-21
Länk till avsnitt

The Impact of Chromatin Modifiers on Disease Development and Progression (Capucine van Rechem)

In this episode of the Epigenetics Podcast, we talked with Capucine van Rechem from Stanford University about her work on the impact of chromatin modifiers on disease development and progression.

During her postdoctoral work, Capucine van Rechem studied the effects of Single nucleotide polymorphisms (SNPs) in KDM4A on lung cancer cell lines and discovered a link between KDM4A and mTOR. She found that cells with the SNP had decreased KDM4A levels and increased sensitivity to inhibitors of the translation pathway. In addition, she found that a combination of histone marks was more predictive of replication timing than RNA expression alone, and identified the specific stages of the cell cycle where KDM4 primarily acts.

Now in her own lab, the focus of her work shifted to SWI-SNF. The team has discovered the role of SWI-SNF in translation through polysome profiling and confirmed the interaction between SWI-SNF and translation. They are currently working to understand the functions of different complexes in translation and their connection to transcription.

 

References

Black, J. C., Manning, A. L., Van Rechem, C., Kim, J., Ladd, B., Cho, J., Pineda, C. M., Murphy, N., Daniels, D. L., Montagna, C., Lewis, P. W., Glass, K., Allis, C. D., Dyson, N. J., Getz, G., & Whetstine, J. R. (2013). KDM4A lysine demethylase induces site-specific copy gain and rereplication of regions amplified in tumors. Cell, 154(3), 541?555. https://doi.org/10.1016/j.cell.2013.06.051

Van Rechem, C., Ji, F., Mishra, S., Chakraborty, D., Murphy, S. E., Dillingham, M. E., Sadreyev, R. I., & Whetstine, J. R. (2020). The lysine demethylase KDM4A controls the cell-cycle expression of replicative canonical histone genes. Biochimica et biophysica acta. Gene regulatory mechanisms, 1863(10), 194624. https://doi.org/10.1016/j.bbagrm.2020.194624

Van Rechem, C., Ji, F., Chakraborty, D., Black, J. C., Sadreyev, R. I., & Whetstine, J. R. (2021). Collective regulation of chromatin modifications predicts replication timing during cell cycle. Cell reports, 37(1), 109799. https://doi.org/10.1016/j.celrep.2021.109799

Ulicna, L., Kimmey, S. C., Weber, C. M., Allard, G. M., Wang, A., Bui, N. Q., Bendall, S. C., Crabtree, G. R., Bean, G. R., & Van Rechem, C. (2022). The Interaction of SWI/SNF with the Ribosome Regulates Translation and Confers Sensitivity to Translation Pathway Inhibitors in Cancers with Complex Perturbations. Cancer research, 82(16), 2829?2837. https://doi.org/10.1158/0008-5472.CAN-21-1360

 

Related Episodes

Oncohistones as Drivers of Pediatric Brain Tumors (Nada Jabado)

H3K4me3, SET Proteins, Isw1, and their Role in Transcription (Jane Mellor)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-09-07
Länk till avsnitt

Long-Range Transcriptional Control by 3D Chromosome Structure (Luca Giorgetti)

In this episode of the Epigenetics Podcast, we caught up with Luca Giorgetti from the Friedrich Miescher Institute to hear about his work on long-range transcriptional control by 3D chromosome structure.

Luca Giorgetti's research focuses on chromosomal interactions, transcriptional output, and the dynamics of enhancer-promoter relationships. His lab investigated the causal relationship between chromosome interactions and transcriptional events. They?ve found that by manipulating the contact probabilities between an enhancer and a promoter by changing their distance, these changes had a substantial effect on transcription levels. This project was an experiment that Luca Giorgetti was eager to do, and it allowed him to establish a smooth functional relationship between contact probabilities and changes in transcription levels.

 

References

Giorgetti, L., Galupa, R., Nora, E. P., Piolot, T., Lam, F., Dekker, J., Tiana, G., & Heard, E. (2014). Predictive polymer modeling reveals coupled fluctuations in chromosome conformation and transcription. Cell, 157(4), 950?963. https://doi.org/10.1016/j.cell.2014.03.025

Redolfi, J., Zhan, Y., Valdes-Quezada, C., Kryzhanovska, M., Guerreiro, I., Iesmantavicius, V., Pollex, T., Grand, R. S., Mulugeta, E., Kind, J., Tiana, G., Smallwood, S. A., de Laat, W., & Giorgetti, L. (2019). DamC reveals principles of chromatin folding in vivo without crosslinking and ligation. Nature structural & molecular biology, 26(6), 471?480. https://doi.org/10.1038/s41594-019-0231-0

Zuin, J., Roth, G., Zhan, Y., Cramard, J., Redolfi, J., Piskadlo, E., Mach, P., Kryzhanovska, M., Tihanyi, G., Kohler, H., Eder, M., Leemans, C., van Steensel, B., Meister, P., Smallwood, S., & Giorgetti, L. (2022). Nonlinear control of transcription through enhancer-promoter interactions. Nature, 604(7906), 571?577. https://doi.org/10.1038/s41586-022-04570-y

Mach, P., Kos, P. I., Zhan, Y., Cramard, J., Gaudin, S., Tünnermann, J., Marchi, E., Eglinger, J., Zuin, J., Kryzhanovska, M., Smallwood, S., Gelman, L., Roth, G., Nora, E. P., Tiana, G., & Giorgetti, L. (2022). Cohesin and CTCF control the dynamics of chromosome folding. Nature genetics, 54(12), 1907?1918. https://doi.org/10.1038/s41588-022-01232-7

 

Related Episodes

scDamID, EpiDamID and Lamina Associated Domains (Jop Kind)

Epigenetics and X-Inactivation (Edith Heard)

Spatial Organization of the Human Genome (Wendy Bickmore)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-08-24
Länk till avsnitt

Transgenerational Inheritance and Epigenetic Imprinting in Plants (Mary Gehring)

In this episode of the Epigenetics Podcast, we talked with Mary Gehring from MIT about her work on transgenerational inheritance and epigenetic imprinting in plants.

Mary Gehring and her team are focusing on plant epigenetics and genetic imprinting in plants, studying DNA methylation in Arabidopsis. They have found significant differences in DNA methylation between the embryo and endosperm of plants, particularly in relation to imprinted genes. She also discusses their work on hydroxymethylcytosine (5-hmC) in Arabidopsis and the challenges of detecting and studying this epigenetic modification.

Next, we discuss the regulatory circuit involving ROS1, a DNA glycosylase involved in demethylation, and its role in maintaining epigenetic homeostasis. The interview concludes with a discussion of CUT&RUN, which the lab has adapted for use in plants. Due to its low input requirements this method has been valuable in studying various plant tissues and has influenced Mary Gehring's research on imprinting in Arabidopsis endosperm.

 

References

Gehring, M., Bubb, K. L., & Henikoff, S. (2009). Extensive demethylation of repetitive elements during seed development underlies gene imprinting. Science (New York, N.Y.), 324(5933), 1447?1451. https://doi.org/10.1126/science.1171609

Pignatta, D., Erdmann, R. M., Scheer, E., Picard, C. L., Bell, G. W., & Gehring, M. (2014). Natural epigenetic polymorphisms lead to intraspecific variation in Arabidopsis gene imprinting. eLife, 3, e03198. https://doi.org/10.7554/eLife.03198

Klosinska, M., Picard, C. L., & Gehring, M. (2016). Conserved imprinting associated with unique epigenetic signatures in the Arabidopsis genus. Nature plants, 2, 16145. https://doi.org/10.1038/nplants.2016.145

Zheng, X. Y., & Gehring, M. (2019). Low-input chromatin profiling in Arabidopsis endosperm using CUT&RUN. Plant reproduction, 32(1), 63?75. https://doi.org/10.1007/s00497-018-00358-1

 

Related Episodes

The Role of Small RNAs in Transgenerational Inheritance in C. elegans (Oded Rechavi)

Epigenetic Influence on Memory Formation and Inheritance (Isabelle Mansuy)

The Epigenetics of Human Sperm Cells (Sarah Kimmins)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-08-10
Länk till avsnitt

When is a Peak a Peak? (Claudio Cantù)

In this episode of the Epigenetics Podcast, we talked to Claudio Cantù from Linköping University about his work on peak blacklists, peak concordance and what is a peak in CUT&RUN.

Our host Stefan Dillinger and guest Claudio Cantù dive into the topic of when we can be sure that a peak is a peak. To help with this, Claudio Cantù's group has been working on defining a set of suspicious peaks that can be used as a "peak blacklist" and can be subtracted to clean up CUT&RUN data sets. The lab also worked on a method called ICEBERG (Increased Capture of Enrichment By Exhaustive Replicate aGgregation) to help define peaks from a number of experimental replicates. By using this algorithm, the team is trying to discover the beta-catenin binding profile, not the tip of the beta-catenin binding iceberg, but the whole of the beta-catenin binding profile.

 

References

Zambanini, G., Nordin, A., Jonasson, M., Pagella, P., & Cantù, C. (2022). A new CUT&RUN low volume-urea (LoV-U) protocol optimized for transcriptional co-factors uncovers Wnt/?-catenin tissue-specific genomic targets. Development (Cambridge, England), 149(23), dev201124. https://doi.org/10.1242/dev.201124

Nordin, A., Zambanini, G., Pagella, P., & Cantù, C. (2022). The CUT&RUN Blacklist of Problematic Regions of the Genome [Preprint]. Genomics. https://doi.org/10.1101/2022.11.11.516118

Nordin, A., Pagella, P., Zambanini, G., & Cantu, C. (2023). Exhaustive identification of genome-wide binding events of transcriptional regulators with ICEBERG [Preprint]. Genomics. https://doi.org/10.1101/2023.06.29.547050

 

Related Episodes

Chromatin Profiling: From ChIP to CUT&RUN, CUT&Tag and CUTAC (Steven Henikoff)

Single Cell Epigenomics in Neuronal Development (Tim Petros)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-07-27
Länk till avsnitt

Analysis of 3D Chromatin Structure Using Super-Resolution Imaging (Alistair Boettiger)

In this episode of the Epigenetics Podcast, we talked with Alistair Boettiger from Stanford University about his work on the analysis of 3D chromatin structure of single cells using super-resolution imaging.

Alistair Boettiger and his team focus on developing advanced microscopy techniques to understand gene regulation at the level of 3D genome organization. They have developed Optical Reconstruction of Chromatin Architecture (ORCA), a microscopy approach to trace the 3-dimensional DNA path in the nucleus with genomic resolution down to 2 kb and a throughput of ~10,000 cells per experiment. These methods enable the identification of structural features with comparable resolution to Hi-C, while the advantages of microscopy such as single cell resolution and multimodal measurements remain.

 

References

Boettiger, A., Bintu, B., Moffitt, J. et al. Super-resolution imaging reveals distinct chromatin folding for different epigenetic states. Nature 529, 418?422 (2016). https://doi.org/10.1038/nature16496

Bogdan Bintu et al., Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 362, eaau1783 (2018). DOI:10.1126/science.aau1783

Mateo, L.J., Sinnott-Armstrong, N. & Boettiger, A.N. Tracing DNA paths and RNA profiles in cultured cells and tissues with ORCA. Nat Protoc 16, 1647?1713 (2021). https://doi.org/10.1038/s41596-020-00478-x

Rajpurkar, A.R., Mateo, L.J., Murphy, S.E. et al. Deep learning connects DNA traces to transcription to reveal predictive features beyond enhancer?promoter contact. Nat Commun 12, 3423 (2021). https://doi.org/10.1038/s41467-021-23831-4

Tzu-Chiao Hung, David M. Kingsley, & Alistair Boettiger. (2023). Boundary stacking interactions enable cross-TAD enhancer-promoter communication during limb development. BioRxiv, 2023.02.06.527380. https://doi.org/10.1101/2023.02.06.527380

Hafner, A., Park, M., Berger, S. E., Murphy, S. E., Nora, E. P., & Boettiger, A. N. (2023). Loop stacking organizes genome folding from TADs to chromosomes. Molecular cell, 83(9), 1377?1392.e6. https://doi.org/10.1016/j.molcel.2023.04.008

 

Related Episodes

Hi-C and Three-Dimensional Genome Sequencing (Erez Lieberman Aiden)

Unraveling Mechanisms of Chromosome Formation (Job Dekker)

Biophysical Modeling of 3-D Genome Organization (Leonid Mirny)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-07-13
Länk till avsnitt

Gene Dosage Alterations in Evolution and Ageing (Claudia Keller Valsecchi)

In this episode of the Epigenetics Podcast, we caught up with Claudia Keller Valsecchi from the Institute for Molecular Biology in Mainz to talk about her work on gene dosage alterations in evolution and ageing.

Claudia Keller-Valsecchi's team focuses on understanding the fundamental mechanisms of how cellular function in eukaryotes is influenced by gene copy number variation. Recent findings indicate that precise MSL2-mediated gene dosage is highly relevant for organismal development. Since 2020 Claudia Keller-Valsecchi runs her own lab at the IMB in Mainz, Germany, where she tries to understand from a molecular mechanistic point of view how gene dosage compensation works, with projects in mosquitoes and in Artemia franciscanagene, as well as dosage regulation in the mammalian system regarding development and disease.

 

References

Keller, C., Adaixo, R., Stunnenberg, R., Woolcock, K. J., Hiller, S., & Bühler, M. (2012). HP1Swi6 Mediates the Recognition and Destruction of Heterochromatic RNA Transcripts. Molecular Cell, 47(2), 215?227. https://doi.org/10.1016/j.molcel.2012.05.009

Valsecchi, C.I.K., Basilicata, M.F., Georgiev, P. et al. RNA nucleation by MSL2 induces selective X chromosome compartmentalization. Nature 589, 137?142 (2021). https://doi.org/10.1038/s41586-020-2935-z

Keller Valsecchi, C. I., Marois, E., Basilicata, M. F., Georgiev, P., & Akhtar, A. (2021). Distinct mechanisms mediate X chromosome dosage compensation in Anopheles and Drosophila. Life Science Alliance, 4(9), e202000996. https://doi.org/10.26508/lsa.202000996

 

Related Episodes

Epigenetics and X-Inactivation (Edith Heard)

Dosage Compensation in Drosophila (Asifa Akhtar)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-06-29
Länk till avsnitt

Structural Analysis of Nucleosomes During Transcription (Lucas Farnung)

In this episode of the Epigenetics Podcast, we caught up with Lucas Farnung from Harvard Medical School to talk about his work on the structural analysis of nucleosomes during transcription.

Lucas Farnung started his scientific career in Patrick Cramer's lab, trying to solve the cryo-EM structure of RNA polymerase II transcribing through a nucleosome. This project spanned some time before being published in 2018, during which time Dr. Farnung accomplished several other goals. The team solved the cryo-electron microscopy structure of Chd1 from the yeast Saccharomyces cerevisiae bound to a nucleosome at a resolution of 4.8 Å, solved the structure of the nucleosome-CHD4 chromatin remodeler, and investigated the structural basis of nucleosome transcription mediated by Chd1 and FACT. In 2021, he started his own lab and is now working on structural analysis of nucleosomes during transcription and how chromatin remodelers work on the chromatin template.

References

Farnung, L., Vos, S. M., Wigge, C., & Cramer, P. (2017). Nucleosome-Chd1 structure and implications for chromatin remodelling. Nature, 550(7677), 539?542. https://doi.org/10.1038/nature24046

Farnung, L., Vos, S. M., & Cramer, P. (2018). Structure of transcribing RNA polymerase II-nucleosome complex. Nature communications, 9(1), 5432. https://doi.org/10.1038/s41467-018-07870-y

Filipovski, M., Soffers, J. H. M., Vos, S. M., & Farnung, L. (2022). Structural basis of nucleosome retention during transcription elongation. Science (New York, N.Y.), 376(6599), 1313?1316. https://doi.org/10.1126/science.abo3851

 

Related Episodes

Molecular Mechanisms of Chromatin Modifying Enzymes (Karim-Jean Armache)

Regulation of Chromatin Organization by Histone Chaperones (Geneviève Almouzni)

Transcription Elongation Control by the Paf1 Complex (Karen Arndt)

From Nucleosome Structure to Function (Karolin Luger)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-06-15
Länk till avsnitt

DNA Methylation Alterations in Neurodegenerative Diseases (Paula Desplats)

In this episode of the Epigenetics Podcast, we caught up with Paula Desplats from the University of California San Diego to talk about her work on DNA Methylation Alterations in Neurodegenerative Diseases.

The laboratory of Paula desalts focuses on decoding the role of epigenetic mechanisms, like DNA methylation, on the onset and progression of neurodegenerative diseases like Parkinson?s and Alzheimer?s. In doing so, on of the goals of the Desplats team is to develop a biomarker panel based on quantification of DNA methylation of selected genes that can discriminate Parkison's Disease patients from healthy subjects in a simple blood test. More recently, the team also focused on the role of the circadian rhythm on neurodegenerative diseases and finding a way how interventions can help in managing the disease.

 

References

Masliah, E., Dumaop, W., Galasko, D., & Desplats, P. (2013). Distinctive patterns of DNA methylation associated with Parkinson disease: identification of concordant epigenetic changes in brain and peripheral blood leukocytes. Epigenetics, 8(10), 1030?1038. https://doi.org/10.4161/epi.25865

Cronin, P., McCarthy, M. J., Lim, A., Salmon, D. P., Galasko, D., Masliah, E., De Jager, P. L., Bennett, D. A., & Desplats, P. (2017). Circadian alterations during early stages of Alzheimer's disease are associated with aberrant cycles of DNA methylation in BMAL1. Alzheimer's & dementia : the journal of the Alzheimer's Association, 13(6), 689?700. https://doi.org/10.1016/j.jalz.2016.10.003

Henderson-Smith, A., Fisch, K. M., Hua, J., Liu, G., Ricciardelli, E., Jepsen, K., Huentelman, M., Stalberg, G., Edland, S. D., Scherzer, C. R., Dunckley, T., & Desplats, P. (2019). DNA methylation changes associated with Parkinson's disease progression: outcomes from the first longitudinal genome-wide methylation analysis in blood. Epigenetics, 14(4), 365?382. https://doi.org/10.1080/15592294.2019.1588682

Nasamran, C. A., Sachan, A., Mott, J., Kuras, Y. I., Scherzer, C. R., Study, H. B., Ricciardelli, E., Jepsen, K., Edland, S. D., Fisch, K. M., & Desplats, P. (2021). Differential blood DNA methylation across Lewy body dementias. Alzheimer's & dementia (Amsterdam, Netherlands), 13(1), e12156. https://doi.org/10.1002/dad2.12156

 

Related Episodes

Development of Integrative Machine Learning Tools for Neurodegenerative Diseases (Enrico Glaab)

The Role of DNA Methylation in Epilepsy (Katja Kobow)

CpG Islands, DNA Methylation, and Disease (Sir Adrian Bird)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-06-01
Länk till avsnitt

scDamID, EpiDamID and Lamina Associated Domains (Jop Kind)

In this episode of the Epigenetics Podcast, we caught up with Jop Kind from Hubrecht Institute to talk about his work on single cell DamID, EpiDamID, and Lamina Associated Domains (LADs).

Jop Kind started out developing single cell DamID (scDamID), based on the DamID technique. First, this technique was adapted to a microscopic readout which enabled them to follow the localisation of chromatin domains after cell division. Next, the lab expanded this technique into the NGS space and created genome-wide maps of nuclear lamina Interactions in single human cells. Since LADs are in a heterochromatic chromatin context, the lab expanded scDamID into the epigenetic space. They first combined it with a transcriptional readout.  Later-on they developed EpiDamID, a method to target a diverse set of chromatin types by taking advantage of the binding specificities of single-chain variable fragment antibodies, engineered chromatin reader domains, and endogenous chromatin-binding proteins.

 

References

Kind, J., Pagie, L., Ortabozkoyun, H., Boyle, S., de Vries, S. S., Janssen, H., Amendola, M., Nolen, L. D., Bickmore, W. A., & van Steensel, B. (2013). Single-Cell Dynamics of Genome-Nuclear Lamina Interactions. Cell, 153(1), 178?192. https://doi.org/10.1016/j.cell.2013.02.028

Kind, J., Pagie, L., de Vries, S. S., Nahidiazar, L., Dey, S. S., Bienko, M., Zhan, Y., Lajoie, B., de Graaf, C. A., Amendola, M., Fudenberg, G., Imakaev, M., Mirny, L. A., Jalink, K., Dekker, J., van Oudenaarden, A., & van Steensel, B. (2015). Genome-wide Maps of Nuclear Lamina Interactions in Single Human Cells. Cell, 163(1), 134?147. https://doi.org/10.1016/j.cell.2015.08.040

Borsos, M., Perricone, S.M., Schauer, T. et al. Genome?lamina interactions are established de novo in the early mouse embryo. Nature 569, 729?733 (2019). https://doi.org/10.1038/s41586-019-1233-0

Markodimitraki, C. M., Rang, F. J., Rooijers, K., de Vries, S. S., Chialastri, A., de Luca, K. L., Lochs, S. J. A., Mooijman, D., Dey, S. S., & Kind, J. (2020). Simultaneous quantification of protein?DNA interactions and transcriptomes in single cells with scDam&T-seq. Nature Protocols, 15(6), 1922?1953. https://doi.org/10.1038/s41596-020-0314-8

Rang, F. J., de Luca, K. L., de Vries, S. S., Valdes-Quezada, C., Boele, E., Nguyen, P. D., Guerreiro, I., Sato, Y., Kimura, H., Bakkers, J., & Kind, J. (2022). Single-cell profiling of transcriptome and histone modifications with EpiDamID. Molecular Cell, 82(10), 1956-1970.e14. https://doi.org/10.1016/j.molcel.2022.03.009

 

Related Episodes

Dosage Compensation in Drosophila (Asifa Akhtar)

Chromatin Profiling: From ChIP to CUT&RUN, CUT&Tag and CUTAC (Steven Henikoff)

Single Cell Epigenomics in Neuronal Development (Tim Petros)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-05-17
Länk till avsnitt

Circulating Epigenetic Biomarkers in Cancer (Charlotte Proudhon)

In this episode of the Epigenetics Podcast, we caught up with Charlotte Proudhon from the Institut Curie to talk about her work on circulating tumor DNA and circulating Epi-mutations as biomarkers in cancer.

Charlotte Proudhon started out her research career by investigating circulating tumor DNA (ctDNA). This kind of DNA is shed into the bloodstream by apoptotic tumor cells and can be analyzed after collection by a simple blood draw, which makes it a very useful biomarker for cancer. Using this approach cancers can be identified by their unique mutational fingerprint. However, soon the limitations of this approach became apparent and the fact that this ctDNA is actually shed into the bloodstream as nucleosomal particles was utilized by the Proudhon team and now the methylation fingerprint of the LINE-1 repeats is used as a biomarker for cancer diagnosis and monitoring of the success of a cancer treatment.

 

References

Decraene, C., Silveira, A. B., Bidard, F. C., Vallée, A., Michel, M., Melaabi, S., Vincent-Salomon, A., Saliou, A., Houy, A., Milder, M., Lantz, O., Ychou, M., Denis, M. G., Pierga, J. Y., Stern, M. H., & Proudhon, C. (2018). Multiple Hotspot Mutations Scanning by Single Droplet Digital PCR. Clinical chemistry, 64(2), 317?328. https://doi.org/10.1373/clinchem.2017.272518

Bortolini Silveira, A., Bidard, F. C., Tanguy, M. L., Girard, E., Trédan, O., Dubot, C., Jacot, W., Goncalves, A., Debled, M., Levy, C., Ferrero, J. M., Jouannaud, C., Rios, M., Mouret-Reynier, M. A., Dalenc, F., Hego, C., Rampanou, A., Albaud, B., Baulande, S., Berger, F., ? Pierga, J. Y. (2021). Multimodal liquid biopsy for early monitoring and outcome prediction of chemotherapy in metastatic breast cancer. NPJ breast cancer, 7(1), 115. https://doi.org/10.1038/s41523-021-00319-4

 

Related Episodes

Epigenome-based Precision Medicine (Eleni Tomazou)

Epigenetics and Epitranscriptomics in Cancer (Manel Esteller)

DNA Methylation and Mammalian Development (Déborah Bourc'his)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-05-04
Länk till avsnitt

Epigenetic Landscapes During Cancer (Luciano Di Croce)

In this episode of the Epigenetics Podcast, we caught up with Luciano Di Croce from the Center of Genomic Regulation in Barcelona to talk about his work on epigenetic landscapes in cancer.

The Di Croce Lab focuses on the Polycomb Complex and its influence on diseases like cancer. Luciano Di Croce started out his research career investigating the oncogenic transcription factor PML-RAR. They could show that in leukemic cells knockdown of SUZ12, a key component of Polycomb repressive complex 2 (PRC2), reverts not only histone modification but also induces DNA de-methylation of PML-RAR target genes. More recently the team focused on two other Polycomb related proteins Zrf1 and PHF19 and were able to characterize some of their functions in gene targeting in different disease and developmental contexts.

 

References

Di Croce, L., Raker, V. A., Corsaro, M., Fazi, F., Fanelli, M., Faretta, M., Fuks, F., Lo Coco, F., Kouzarides, T., Nervi, C., Minucci, S., & Pelicci, P. G. (2002). Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science (New York, N.Y.), 295(5557), 1079?1082. https://doi.org/10.1126/science.1065173

Richly, H., Rocha-Viegas, L., Ribeiro, J. D., Demajo, S., Gundem, G., Lopez-Bigas, N., Nakagawa, T., Rospert, S., Ito, T., & Di Croce, L. (2010). Transcriptional activation of polycomb-repressed genes by ZRF1. Nature, 468(7327), 1124?1128. https://doi.org/10.1038/nature09574

Jain, P., Ballare, C., Blanco, E., Vizan, P., & Di Croce, L. (2020). PHF19 mediated regulation of proliferation and invasiveness in prostate cancer cells. eLife, 9, e51373. https://doi.org/10.7554/eLife.51373

 

Related Episodes

Oncohistones as Drivers of Pediatric Brain Tumors (Nada Jabado)

Transcription and Polycomb in Inheritance and Disease (Danny Reinberg)

Targeting COMPASS to Cure Childhood Leukemia (Ali Shilatifard)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-04-20
Länk till avsnitt

Formation of CenH3-deficient Kinetochores (Ines Drinnenberg)

In this episode of the Epigenetics Podcast, we caught up with Ines Drinnenberg from Institute Curie to talk about her work on the formation of CenH3-deficient kinetochores.

The laboratory of Ines Drinneberg focuses on centromeres and how different strategies of centromere organization have evolved in different organisms. While most eukaryotes have monocentric chromosomes, where spindle attachment is restricted to a single chromosomal region resembling such classic X-shape like structures under the microscope, many lineages have evolved holocentric chromosomes where spindle microtubules attach along the entire length of the chromosome. The team was able to show the independent loss of CENH3/CENP-A in holocentric insects. Furthermore, the team focuses on how CenH3-deficient kinetochores form and were able to identify several conserved kinetochore components that emerged as a key component for CenH3-deficient kinetochore formation in Lepidoptera.

 

References

Drinnenberg, I. A., deYoung, D., Henikoff, S., & Malik, H. S. (2014). Recurrent loss of CenH3 is associated with independent transitions to holocentricity in insects. eLife, 3, e03676. https://doi.org/10.7554/eLife.03676

Molaro, A., & Drinnenberg, I. A. (2018). Studying the Evolution of Histone Variants Using Phylogeny. Methods in molecular biology (Clifton, N.J.), 1832, 273?291. https://doi.org/10.1007/978-1-4939-8663-7_15

Cortes-Silva, N., Ulmer, J., Kiuchi, T., Hsieh, E., Cornilleau, G., Ladid, I., Dingli, F., Loew, D., Katsuma, S., & Drinnenberg, I. A. (2020). CenH3-Independent Kinetochore Assembly in Lepidoptera Requires CCAN, Including CENP-T. Current biology : CB, 30(4), 561?572.e10. https://doi.org/10.1016/j.cub.2019.12.014

Senaratne, A. P., Muller, H., Fryer, K. A., Kawamoto, M., Katsuma, S., & Drinnenberg, I. A. (2021). Formation of the CenH3-Deficient Holocentromere in Lepidoptera Avoids Active Chromatin. Current biology : CB, 31(1), 173?181.e7. https://doi.org/10.1016/j.cub.2020.09.078

Vanpoperinghe, L., Carlier-Grynkorn, F., Cornilleau, G., Kusakabe, T., Drinnenberg, I. A., & Tran, P. T. (2021). Live-cell imaging reveals square shape spindles and long mitosis duration in the silkworm holocentric cells. microPublication biology, 2021, 10.17912/micropub.biology.000441. https://doi.org/10.17912/micropub.biology.000441

 

Related Episodes

The Role of Non-Histone Proteins in Chromosome Structure and Function During Mitosis (Bill Earnshaw)

Chromatin Profiling: From ChIP to CUT&RUN, CUT&Tag and CUTAC (Steven Henikoff)

In Vivo Nucleosome Structure and Dynamics (Srinivas Ramachandran)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-04-06
Länk till avsnitt

Effects of Environmental Cues on the Epigenome and Longevity (Paul Shiels)

In this episode of the Epigenetics Podcast, we caught up with Paul Shiels from the University of Glasgow to talk about his work on the effects of environmental cues on the epigenome and longevity.

Paul Shiels and his team focus on the question on how age related health is influenced by the environment. Factors like the socio-economic position, nutrition, lifestyle and the environment can influence the microbiome and the inflammation burden on the body which in turn can alter individual trajectories of ageing and health. The lab also tries to understand the epigenetic, molecular and cellular mechanisms that link the exposome to chronic age related diseases of older people. They have shown that (1)  imbalanced nutrition is associated with a microbiota-mediated accelerated ageing in the general population, (2) a significantly higher abundance of circulatory pathogenic bacteria is found in the most biologically aged, while those less biologically aged possess more circulatory salutogenic bacteria with a capacity to metabolise and produce cytoprotective Nrf2 agonists, (3) those at lower socioeconomic position possess significantly lower betaine levels indicative of a poorer diet and poorer health span and consistent with reduced global DNA methylation levels in this group.

 

References

Harris, S. E., Deary, I. J., MacIntyre, A., Lamb, K. J., Radhakrishnan, K., Starr, J. M., Whalley, L. J., & Shiels, P. G. (2006). The association between telomere length, physical health, cognitive ageing, and mortality in non-demented older people. Neuroscience Letters, 406(3), 260?264. https://doi.org/10.1016/j.neulet.2006.07.055

Paul G. Shiels, Improving Precision in Investigating Aging: Why Telomeres Can Cause Problems, The Journals of Gerontology: Series A, Volume 65A, Issue 8, August 2010, Pages 789?791, https://doi.org/10.1093/gerona/glq095

Mafra D, Ugochukwu SA, Borges NA, et al. Food for healthier aging: power on your plate. Critical Reviews in Food Science and Nutrition. 2022 Aug:1-14. DOI: 10.1080/10408398.2022.2107611. PMID: 35959705.

Shiels PG, Stenvinkel P, Kooman JP, McGuinness D. Circulating markers of ageing and allostatic load: A slow train coming. Practical Laboratory Medicine. 2017 Apr;7:49-54. DOI: 10.1016/j.plabm.2016.04.002. PMID: 28856219; PMCID: PMC5574864.

 

Related Episodes

Transposable Elements in Gene Regulation and Evolution (Marco Trizzino)

Epigenetic Clocks and Biomarkers of Ageing (Morgan Levine)

Aging and Epigenetics (Peter Tessarz)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-03-23
Länk till avsnitt

The Epigenetics of Human Sperm Cells (Sarah Kimmins)

In this episode of the Epigenetics Podcast, we caught up with Sarah Kimmins from Université de Montreal to talk about her work on the epigenetics of human sperm cells.

The focus of Sarah Kimmins and her lab is how sperm and offspring health is impacted by the father's environment. The core of this is the sperm epigenome, which has been implicated in complex diseases such as infertility, cancer, diabetes, schizophrenia and autism. The Kimmins lab is interested which players play a role in this and came across the Histone post-translational modification H3K4me3. In this interview we talk about how the father's life choices can impact offspring health, which can also be inherited transgenerationally and how this can be used to develop intervention strategies to improve child and adult health.

 

References

Siklenka, K., Erkek, S., Godmann, M., Lambrot, R., McGraw, S., Lafleur, C., Cohen, T., Xia, J., Suderman, M., Hallett, M., Trasler, J., Peters, A. H., & Kimmins, S. (2015). Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science (New York, N.Y.), 350(6261), aab2006. https://doi.org/10.1126/science.aab2006

Lismer, A., Siklenka, K., Lafleur, C., Dumeaux, V., & Kimmins, S. (2020). Sperm histone H3 lysine 4 trimethylation is altered in a genetic mouse model of transgenerational epigenetic inheritance. Nucleic acids research, 48(20), 11380?11393. https://doi.org/10.1093/nar/gkaa712

Lismer, A., Dumeaux, V., Lafleur, C., Lambrot, R., Brind'Amour, J., Lorincz, M. C., & Kimmins, S. (2021). Histone H3 lysine 4 trimethylation in sperm is transmitted to the embryo and associated with diet-induced phenotypes in the offspring. Developmental cell, 56(5), 671?686.e6. https://doi.org/10.1016/j.devcel.2021.01.014

 

Related Episodes

H3K4me3, SET Proteins, Isw1, and their Role in Transcription (Jane Mellor)

The Effects of Early Life Stress on Mammalian Development (Catherine J. Peña)

DNA Methylation and Mammalian Development (Déborah Bourc'his)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-03-09
Länk till avsnitt

Transgenerational Inheritance and Evolution of Epimutations (Peter Sarkies)

In this episode of the Epigenetics Podcast, we caught up with Peter Sarkies from University of Oxford Biochemistry to talk about his work on Transgenerational Inheritance of Epimutations.

The team in the Sarkies lab focuses on investigating the connections between epigenetic gene regulation and evolution. The lab performs evolution experiments in the nematode C. elegans to determine if evolution can be influenced by epigenetic differences between individuals in a given population when no changes in the underlying DNA sequence are observed. A second area of interest of the team is evolution of piRNAs, which are present in metazoans but have been lost in nematodes during evolution.

 

References

The Selfish Gene

Sarkies, P., & Miska, E. A. (2013). Is There Social RNA? Science, 341(6145), 467?468. https://doi.org/10.1126/science.1243175

Beltran, T., Shahrezaei, V., Katju, V., & Sarkies, P. (2020). Epimutations driven by small RNAs arise frequently but most have limited duration in Caenorhabditis elegans. Nature ecology & evolution, 4(11), 1539?1548. https://doi.org/10.1038/s41559-020-01293-z

Beltran, T., Pahita, E., Ghosh, S., Lenhard, B., & Sarkies, P. (2021). Integrator is recruited to promoter-proximally paused RNA Pol II to generate Caenorhabditis elegans piRNA precursors. The EMBO journal, 40(5), e105564. https://doi.org/10.15252/embj.2020105564

 

Related Episodes

The Role of Small RNAs in Transgenerational Inheritance in C. elegans (Oded Rechavi)

Epigenetic Influence on Memory Formation and Inheritance (Isabelle Mansuy)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-02-23
Länk till avsnitt

Transcription Elongation Control by the Paf1 Complex (Karen Arndt)

In this episode of the Epigenetics Podcast, we caught up with Karen Arndt from the University of Pittsburgh to talk about her work on transcription elongation control by the Paf1 complex.

Karen Arndt and her lab investigate the process of transcriptional elongation and how RNA polymerase II overcomes obstacles like nucleosomes. One of the proteins that helps overcome those obstacles is the Paf1 complex. This complex associates with the transcribing polymerase and helps in modifying the chromatin template by ubiquitinating Histone H2B and methylating Histone H3.

 

References

Squazzo, S. L., Costa, P. J., Lindstrom, D. L., Kumer, K. E., Simic, R., Jennings, J. L., Link, A. J., Arndt, K. M., & Hartzog, G. A. (2002). The Paf1 complex physically and functionally associates with transcription elongation factors in vivo. The EMBO journal, 21(7), 1764?1774. https://doi.org/10.1093/emboj/21.7.1764

Van Oss, S. B., Shirra, M. K., Bataille, A. R., Wier, A. D., Yen, K., Vinayachandran, V., Byeon, I. L., Cucinotta, C. E., Héroux, A., Jeon, J., Kim, J., VanDemark, A. P., Pugh, B. F., & Arndt, K. M. (2016). The Histone Modification Domain of Paf1 Complex Subunit Rtf1 Directly Stimulates H2B Ubiquitylation through an Interaction with Rad6. Molecular cell, 64(4), 815?825. https://doi.org/10.1016/j.molcel.2016.10.008

Cucinotta, C. E., Hildreth, A. E., McShane, B. M., Shirra, M. K., & Arndt, K. M. (2019). The nucleosome acidic patch directly interacts with subunits of the Paf1 and FACT complexes and controls chromatin architecture in vivo. Nucleic acids research, 47(16), 8410?8423. https://doi.org/10.1093/nar/gkz549

Hildreth, A. E., Ellison, M. A., Francette, A. M., Seraly, J. M., Lotka, L. M., & Arndt, K. M. (2020). The nucleosome DNA entry-exit site is important for transcription termination and prevention of pervasive transcription. eLife, 9, e57757. https://doi.org/10.7554/eLife.57757

 

Related Episodes

Targeting COMPASS to Cure Childhood Leukemia (Ali Shilatifard)

H3K4me3, SET Proteins, Isw1, and their Role in Transcription (Jane Mellor)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-02-09
Länk till avsnitt

Molecular Mechanisms of Chromatin Modifying Enzymes (Karim-Jean Armache)

In this episode of the Epigenetics Podcast, we caught up with Karim-Jean Armache from New York University - Grossman School of Medicine to talk about his work on the structural analysis of Polycomb Complex Proteins and molecular mechanisms of chromatin modifying enzymes.

Karim-Jean Armache started his research career with the structural characterization of the 12-subunit RNA Polymerase II. After starting his own lab he used this knowledge in x-ray crystallography and electron microscopy to study how gene silencing complexes like the PRC complex act on chromatin and influence transcription. Further work in the Armache Lab focused on Dot, a  histone H3K79 methyltransferase, and how it acts on chromatin, as well as how it is regulated by Histone-Histone crosstalk.

References

Armache, K. J., Garlick, J. D., Canzio, D., Narlikar, G. J., & Kingston, R. E. (2011). Structural basis of silencing: Sir3 BAH domain in complex with a nucleosome at 3.0 Å resolution. Science (New York, N.Y.), 334(6058), 977?982. https://doi.org/10.1126/science.1210915

Lee, C. H., Holder, M., Grau, D., Saldaña-Meyer, R., Yu, J. R., Ganai, R. A., Zhang, J., Wang, M., LeRoy, G., Dobenecker, M. W., Reinberg, D., & Armache, K. J. (2018). Distinct Stimulatory Mechanisms Regulate the Catalytic Activity of Polycomb Repressive Complex 2. Molecular cell, 70(3), 435?448.e5. https://doi.org/10.1016/j.molcel.2018.03.019

De Ioannes, P., Leon, V. A., Kuang, Z., Wang, M., Boeke, J. D., Hochwagen, A., & Armache, K. J. (2019). Structure and function of the Orc1 BAH-nucleosome complex. Nature communications, 10(1), 2894. https://doi.org/10.1038/s41467-019-10609-y

Valencia-Sánchez, M. I., De Ioannes, P., Wang, M., Truong, D. M., Lee, R., Armache, J. P., Boeke, J. D., & Armache, K. J. (2021). Regulation of the Dot1 histone H3K79 methyltransferase by histone H4K16 acetylation. Science (New York, N.Y.), 371(6527), eabc6663. https://doi.org/10.1126/science.abc6663

 

Related Episodes

Transcription and Polycomb in Inheritance and Disease (Danny Reinberg)

From Nucleosome Structure to Function (Karolin Luger)

Oncohistones as Drivers of Pediatric Brain Tumors (Nada Jabado)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

eMail: [email protected]

2023-01-26
Länk till avsnitt

The Role of PHF13 in Chromatin and Transcription (Sarah Kinkley)

In this episode of the Epigenetics Podcast, we caught up with Sarah Kinkley from the Max Planck Institute of Molecular Genetics to talk about her work on PHF13 and its role in chromatin and transcription.

The Kinkley laboratory focuses mainly on unraveling the mechanism of action of the transcription factor PHF13 (PHC Finger Protein 13). PHF13 is a reader of the epigenetic mark H3K4 trimethylation which influences higher chromatin order, transcriptional regulation, and differentiation. The lab has shown that PHF13 plays a crucial role in phase separation and mitotic chromatin compaction.

 

References

Kinkley, S., Staege, H., Mohrmann, G., Rohaly, G., Schaub, T., Kremmer, E., Winterpacht, A., & Will, H. (2009). SPOC1: a novel PHD-containing protein modulating chromatin structure and mitotic chromosome condensation. Journal of cell science, 122(Pt 16), 2946?2956. https://doi.org/10.1242/jcs.047365

Chung, H. R., Xu, C., Fuchs, A., Mund, A., Lange, M., Staege, H., Schubert, T., Bian, C., Dunkel, I., Eberharter, A., Regnard, C., Klinker, H., Meierhofer, D., Cozzuto, L., Winterpacht, A., Di Croce, L., Min, J., Will, H., & Kinkley, S. (2016). PHF13 is a molecular reader and transcriptional co-regulator of H3K4me2/3. eLife, 5, e10607. https://doi.org/10.7554/eLife.10607

Connecting the Dots: PHF13 and cohesin promote polymer-polymer phase separation of chromatin into chromosomes. Francesca Rossi, Rene Buschow, Laura V. Glaser, Tobias Schubert, Hannah Staege, Astrid Grimme, Hans Will, Thorsten Milke, Martin Vingron, Andrea M. Chiariello, Sarah Kinkley. bioRxiv 2022.03.04.482956; doi: https://doi.org/10.1101/2022.03.04.482956

 

Related Episodes

The Role of Blimp-1 in Immune-Cell Differentiation (Erna Magnúsdóttir)

H3K4me3, SET Proteins, Isw1, and their Role in Transcription (Jane Mellor)

The Role of SMCHD1 in Development and Disease (Marnie Blewitt)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2023-01-12
Länk till avsnitt

Genome Organization Mediated by RNA Polymerase II (Argyrys Papantonis)

In this episode of the Epigenetics Podcast, we caught up with Akis Papantonis from the University Medical Center Göttingen to talk about his work on genome organisation mediated by RNA Polymerase II.

The research of the Papantonis Laboratory focuses on investigating how environmental signalling stimuli are integrated by chromatin to control homeostatic to deregulated functional transitions. In more detail, the team is interested in how dynamic higher-order regulatory networks are influenced by the underlying linear DNA fiber. The ultimate goal of the laboratory is to understand general rules governing transcriptional and chromatin homeostasis and finally, how those rules might affect development, ageing or malignancies.

 

References

Larkin, J. D., Cook, P. R., & Papantonis, A. (2012). Dynamic reconfiguration of long human genes during one transcription cycle. Molecular and cellular biology, 32(14), 2738?2747. https://doi.org/10.1128/MCB.00179-12

Diermeier, S., Kolovos, P., Heizinger, L., Schwartz, U., Georgomanolis, T., Zirkel, A., Wedemann, G., Grosveld, F., Knoch, T. A., Merkl, R., Cook, P. R., Längst, G., & Papantonis, A. (2014). TNF? signalling primes chromatin for NF-?B binding and induces rapid and widespread nucleosome repositioning. Genome biology, 15(12), 536. https://doi.org/10.1186/s13059-014-0536-6

Sofiadis, K., Josipovic, N., Nikolic, M., Kargapolova, Y., Übelmesser, N., Varamogianni-Mamatsi, V., Zirkel, A., Papadionysiou, I., Loughran, G., Keane, J., Michel, A., Gusmao, E. G., Becker, C., Altmüller, J., Georgomanolis, T., Mizi, A., & Papantonis, A. (2021). HMGB1 coordinates SASP-related chromatin folding and RNA homeostasis on the path to senescence. Molecular systems biology, 17(6), e9760. https://doi.org/10.15252/msb.20209760

Enhancer-promoter contact formation requires RNAPII and antagonizes loop extrusion. Shu Zhang, Nadine Übelmesser, Mariano Barbieri, Argyris Papantonis. bioRxiv 2022.07.04.498738; doi: https://doi.org/10.1101/2022.07.04.498738

 

Related Episodes

Chromatin Organization During Development and Disease (Marieke Oudelaar)

Biophysical Modeling of 3-D Genome Organization (Leonid Mirny)

Hi-C and Three-Dimensional Genome Sequencing (Erez Lieberman Aiden)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2022-12-15
Länk till avsnitt

The Role of Pioneer Factors Zelda and Grainyhead at the Maternal-to-Zygotic Transition (Melissa Harrison)

In this episode of the Epigenetics Podcast, we caught up with Melissa Harrison from the University of Wisconsin-Madison to talk about her work on the ?Pioneer? Transcription Factors - Zelda and Grainyhead - and their role at the maternal-to-zygotic transition.

The Harrison lab studies how differentiation and development are driven by coordinated changes in gene expression. To do this, the targets of choice are the transcription factors Zelda and Grainyhead that bind to the genome at specific and crucial points in development and differentiation. These specialised transcription factors have the ability to bind to DNA in the context of nucleosomes which defines regulatory elements and leads to subsequent binding of additional classical transcription factors. These properties allow pioneer factors to act at the top of gene regulatory networks and control developmental transitions.

 

References

Harrison, M. M., Botchan, M. R., & Cline, T. W. (2010). Grainyhead and Zelda compete for binding to the promoters of the earliest-expressed Drosophila genes. Developmental biology, 345(2), 248?255. https://doi.org/10.1016/j.ydbio.2010.06.026

Harrison, M. M., Li, X. Y., Kaplan, T., Botchan, M. R., & Eisen, M. B. (2011). Zelda binding in the early Drosophila melanogaster embryo marks regions subsequently activated at the maternal-to-zygotic transition. PLoS genetics, 7(10), e1002266. https://doi.org/10.1371/journal.pgen.1002266

McDaniel, S. L., Gibson, T. J., Schulz, K. N., Fernandez Garcia, M., Nevil, M., Jain, S. U., Lewis, P. W., Zaret, K. S., & Harrison, M. M. (2019). Continued Activity of the Pioneer Factor Zelda Is Required to Drive Zygotic Genome Activation. Molecular cell, 74(1), 185?195.e4. https://doi.org/10.1016/j.molcel.2019.01.014

McDaniel, S. L., & Harrison, M. M. (2019). Optogenetic Inactivation of Transcription Factors in the Early Embryo of Drosophila. Bio-protocol, 9(13), e3296. https://doi.org/10.21769/BioProtoc.3296

Larson, E.D., Komori, H., Gibson, T.J. et al. Cell-type-specific chromatin occupancy by the pioneer factor Zelda drives key developmental transitions in Drosophila. Nat Commun 12, 7153 (2021). https://doi.org/10.1038/s41467-021-27506-y

 

Related Episodes

Pioneer Transcription Factors and Their Influence on Chromatin Structure (Ken Zaret)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2022-12-01
Länk till avsnitt

Epigenetics in Human Malaria Parasites (Elena Gómez-Diaz)

In this episode of the Epigenetics Podcast, we caught up with Elena Gomez-Diaz from the Institute of Parasitology and Biomedicine López-Neyra at the Spanish National Research Council. She share with us her work on the Epigenetics in Human Malaria Parasites.

Elena Gómez-Díaz and her team are focusing on understanding how epigenetic processes are implicated in host-parasite interactions by regulating gene expression in the model of malaria. The team has started to investigate and uncover layers of chromatin regulation that control developmental transitions in Plasmodium falciparum, especially in the parts of the life cycle that take place in the mosquito. Furthermore, the lab has investigated epigenetic changes that are present in malaria-infected Anopheles mosquitos, this led to the identification of cis-regulatory elements and enhancer-promoter networks in response to infection.

 

References

Gómez-Díaz E, Rivero A, Chandre F, Corces VG. Insights into the epigenomic landscape of the human malaria vector Anopheles gambiae. Front Genet. 2014 Aug 15;5:277. doi: 10.3389/fgene.2014.00277. PMID: 25177345; PMCID: PMC4133732.

Gómez-Díaz, E., Yerbanga, R., Lefèvre, T. et al. Epigenetic regulation of Plasmodium falciparum clonally variant gene expression during development in Anopheles gambiae. Sci Rep 7, 40655 (2017). https://doi.org/10.1038/srep40655

José Luis Ruiz, Juan J Tena, Cristina Bancells, Alfred Cortés, José Luis Gómez-Skarmeta, Elena Gómez-Díaz, Characterization of the accessible genome in the human malaria parasite. Plasmodium falciparum, Nucleic Acids Research, Volume 46, Issue 18, 12 October 2018, Pages 9414?9431, https://doi.org/10.1093/nar/gky643

Women in Malaria 2021: A Conference Premier. (2021). Trends in Parasitology, 37(7), 573?580. https://doi.org/10.1016/j.pt.2021.04.001

Twitter Account: https://twitter.com/womeninmalaria

 

Related Episodes

Multiple challenges of CUT&Tag (Cassidee McDonough, Kyle Tanguay)

ATAC-Seq, scATAC-Seq and Chromatin Dynamics in Single-Cells (Jason Buenrostro)

 

Contact

Epigenetics Podcast on Twitter

Epigenetics Podcast on Instagram

Epigenetics Podcast on Mastodon

Active Motif on Twitter

Active Motif on LinkedIn

Email: [email protected]

2022-11-17
Länk till avsnitt

Bioinformatic Analysis in Epigenetics Research (Nick Pervolarakis)

In this episode of the Epigenetics Podcast, we caught up with Nick Pervolarakis from Active Motif to talk about bioinformatic analysis in epigenetics research.

While many ?bench scientists? are familiar with the workflows of ChIP-Seq, ATAC-Seq and CUT&Tag, and even the preparation and analysis of the libraries, the steps between sequencing and fully analyzed data is sometimes thought of as a mystery known only to bioinformatic experts. Most of us have some understanding that the raw data is usually in a file format called a FASTQ. But how do we get from FASTQ files to peaks on a genome browser? This Podcast Episode will provide a peek behind the curtain of the informatic analysis we perform at Active Motif, as part of our end-to-end epigenetic services.

 

References

Life in the FASTQ Lane

Bioinformatics Resource Center

Epigenetic Services

 

Related Episodes

Multiple challenges of ATAC-Seq, Points to Consider (Yuan Xue)

Multiple challenges of CUT&Tag (Cassidee McDonough, Kyle Tanguay)

Multiple Challenges in ChIP (Adam Blattler)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

c8dqseb1h2Zyf2blC15qkyPgu9sB5stin27nqhzP

2022-11-03
Länk till avsnitt

Anchor-Based Bisulfite Sequencing (Ben Delatte)

In this episode of the Epigenetics Podcast, we caught up with Ben Delatte Research Scientist at Active Motif to talk about his work on Anchor Based Bisulfite Sequencing.

Whole Genome Bisulfite Sequencing (WGBS) is the current standard for DNA methylation profiling. However, this approach is costly as it requires sequencing coverage over the entire genome. Here we introduce Anchor-Based Bisulfite Sequencing (ABBS). ABBS captures accurate DNA methylation information in Escherichia coli and mammals, while requiring up to 10 times fewer sequencing reads than WGBS. ABBS interrogates the entire genome and is not restricted to the CpG islands assayed by methods like Reduced Representation Bisulfite Sequencing (RRBS). The ABBS protocol is simple and can be performed in a single day.

 

References

Chapin, N., Fernandez, J., Poole, J. et al. Anchor-based bisulfite sequencing determines genome-wide DNA methylation. Commun Biol 5, 596 (2022). https://doi.org/10.1038/s42003-022-03543-1

 

Related Episodes

The Role of DNA Methylation in Epilepsy (Katja Kobow)

DNA Methylation and Mammalian Development (Déborah Bourc'his)

Effects of DNA Methylation on Diabetes (Charlotte Ling)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-10-20
Länk till avsnitt

Enhancer Communities in Adipocyte Differentiation (Susanne Mandrup)

In this episode of the Epigenetics Podcast, we caught up with Susanne Mandrup from the University of Southern Denmark to talk about her work on the role of enhancer communities in adipocyte differentiation.

The Laboratory of Susanne Mandrup focuses on the effect of enhancers and enhancer communities on the differentiation of mesenchymal stem cell into adipocytes and osteoblasts. The team has shown that there is significant cross-talk between enhancers and that these form communities of highly interconnected enhancers. Inactive enhancers are then activated by association with these pre-existing enhancer networks to facilitate gene expression in adipocyte differentiation.

 

References

Siersbæk R, Rabiee A, Nielsen R, Sidoli S, Traynor S, Loft A, Poulsen LC, Rogowska-Wrzesinska A, Jensen ON, Mandrup S. Transcription factor cooperativity in early adipogenic hotspots and super-enhancers. Cell Rep. 2014 Jun 12;7(5):1443-1455. doi: 10.1016/j.celrep.2014.04.042. Epub 2014 May 22. PMID: 24857652.

Siersbæk R, Baek S, Rabiee A, Nielsen R, Traynor S, Clark N, Sandelin A, Jensen ON, Sung MH, Hager GL, Mandrup S. Molecular architecture of transcription factor hotspots in early adipogenesis. Cell Rep. 2014 Jun 12;7(5):1434-1442. doi: 10.1016/j.celrep.2014.04.043. Epub 2014 May 22. PMID: 24857666; PMCID: PMC6360525.

Siersbæk R, Madsen JGS, Javierre BM, Nielsen R, Bagge EK, Cairns J, Wingett SW, Traynor S, Spivakov M, Fraser P, Mandrup S. Dynamic Rewiring of Promoter-Anchored Chromatin Loops during Adipocyte Differentiation. Mol Cell. 2017 May 4;66(3):420-435.e5. doi: 10.1016/j.molcel.2017.04.010. PMID: 28475875.

Rauch, A., Haakonsson, A.K., Madsen, J.G.S. et al. Osteogenesis depends on commissioning of a network of stem cell transcription factors that act as repressors of adipogenesis. Nat Genet 51, 716?727 (2019). https://doi.org/10.1038/s41588-019-0359-1

Madsen, J.G.S., Madsen, M.S., Rauch, A. et al. Highly interconnected enhancer communities control lineage-determining genes in human mesenchymal stem cells. Nat Genet 52, 1227?1238 (2020). https://doi.org/10.1038/s41588-020-0709-z

 

Related Episodes

Ultraconserved Enhancers and Enhancer Redundancy (Diane Dickel)

Effects of DNA Methylation on Diabetes (Charlotte Ling)

Epigenetic Regulation of Stem Cell Self-Renewal and Differentiation (Peggy Goodell)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-10-06
Länk till avsnitt

Transposable Elements in Gene Regulation and Evolution (Marco Trizzino)

In this episode of the Epigenetics Podcast, we caught up with Marco Trizzino from Thomas Jefferson University to talk about his work on transposable elements in gene regulation and evolution.

Marco Trizzino and his team focus on characterising transposable elements and how they affect gene regulation, evolution and ageing in primates. They could show that transposable elements that integrated into the genome turned into regulatory elements in the genome, like enhancers. They then contribute to regulation of processes like development or ageing, which could be among those factors that lead to increased brain development or longevity in great apes.

 

References

Trizzino M, Park Y, Holsbach-Beltrame M, Aracena K, Mika K, Caliskan M, Perry GH, Lynch VJ, Brown CD. Transposable elements are the primary source of novelty in primate gene regulation. Genome Res. 2017 Oct;27(10):1623-1633. doi: 10.1101/gr.218149.116. Epub 2017 Aug 30. PMID: 28855262; PMCID: PMC5630026.

Pagliaroli L, Porazzi P, Curtis AT, Scopa C, Mikkers HMM, Freund C, Daxinger L, Deliard S, Welsh SA, Offley S, Ott CA, Calabretta B, Brugmann SA, Santen GWE, Trizzino M. Inability to switch from ARID1A-BAF to ARID1B-BAF impairs exit from pluripotency and commitment towards neural crest formation in ARID1B-related neurodevelopmental disorders. Nat Commun. 2021 Nov 9;12(1):6469. doi: 10.1038/s41467-021-26810-x. PMID: 34753942; PMCID: PMC8578637.

Tejada-Martinez D, Avelar RA, Lopes I, Zhang B, Novoa G, de Magalhães JP, Trizzino M. Positive Selection and Enhancer Evolution Shaped Lifespan and Body Mass in Great Apes. Mol Biol Evol. 2022 Feb 3;39(2):msab369. doi: 10.1093/molbev/msab369. PMID: 34971383; PMCID: PMC8837823.

Young transposable elements rewired gene regulatory networks in human and chimpanzee hippocampal intermediate progenitors. Sruti Patoori, Samantha M. Barnada, Christopher Large, John I. Murray, Marco Trizzino. bioRxiv 2021.11.24.469877; doi: https://doi.org/10.1101/2021.11.24.469877

 

Related Episodes

Enhancer-Promoter Interactions During Development (Yad Ghavi-Helm)

Chromatin Organization During Development and Disease (Marieke Oudelaar)

Ultraconserved Enhancers and Enhancer Redundancy (Diane Dickel)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-09-22
Länk till avsnitt

Hydroxymethylation Landscape in Immunecells (Marcela Sjöberg)

In this episode of the Epigenetics Podcast, we caught up with Marcela Sjöberg from the Pontificia Universidad Católica de Chile to talk about her work on the hydroxymethylation landscape in immune cells.

At the beginning of her career Marcela Sjöberg worked on Aurora B and Polycomb and how modifications placed by them modulate the binding of RNA Pol II. Later, her focus shifted to examine cytosine DNA methylation and hydroxymethylation changes in immune cells and how the epigenetic state of these marks varies between individuals and is reprogrammed for Metastable Epialleles in mouse. More recently, the laboratory is interested on how hydroxymethylation of transcription factor binding motifs influence binding and activity of the respective transcription factors in immune cells.

 

References

Sabbattini, P., Sjoberg, M., Nikic, S., Frangini, A., Holmqvist, P.-H., Kunowska, N., Carroll, T., Brookes, E., Arthur, S. J., Pombo, A., & Dillon, N. (2014). An H3K9/S10 methyl-phospho switch modulates Polycomb and Pol II binding at repressed genes during differentiation. Molecular Biology of the Cell, 25(6), 904?915. https://doi.org/10.1091/mbc.e13-10-0628

Kazachenka, A., Bertozzi, T. M., Sjoberg-Herrera, M. K., Walker, N., Gardner, J., Gunning, R., Pahita, E., Adams, S., Adams, D., & Ferguson-Smith, A. C. (2018). Identification, Characterization, and Heritability of Murine Metastable Epialleles: Implications for Non-genetic Inheritance. Cell, 175(5), 1259-1271.e13. https://doi.org/10.1016/j.cell.2018.09.043

Westoby, J., Herrera, M.S., Ferguson-Smith, A.C. et al. Simulation-based benchmarking of isoform quantification in single-cell RNA-seq. Genome Biol 19, 191 (2018). https://doi.org/10.1186/s13059-018-1571-5

Viner, C., Johnson, J., Walker, N., Shi, H., Sjöberg, M., Adams, D. J., Ferguson-Smith, A. C., Bailey, T. L., & Hoffman, M. M. (2016). Modeling methyl-sensitive transcription factor motifs with an expanded epigenetic alphabet [Preprint]. Bioinformatics. https://doi.org/10.1101/043794

 

Related Episodes

The Role of DNA Methylation in Epilepsy (Katja Kobow)

DNA Methylation and Mammalian Development (Déborah Bourc'his)

Effects of DNA Methylation on Chromatin Structure and Transcription (Dirk Schübeler)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-09-08
Länk till avsnitt

Single Cell Epigenomics in Neuronal Development (Tim Petros)

In this episode of the Epigenetics Podcast, we caught up with Tim Petros from the Eunice Kennedy Shriver National Institute of Child Health and Human Development at the NIH to talk about his work on Single Cell Epigenomics in Neuronal Development. 

The Petros lab focuses on ?interneurons?, their diversity and how environmental signals interact to generate this diversity. This subgroup of neurons comprise about 20% of neutrons in the brain, however, they are the primary source of inhibition. Furthermore, interneurons are critical components in modulating information flow throughout the nervous system. The Petros lab seeks to uncover the genetic programs that lead to the incredible diversity in interneurons, as well as how the local environment influences this process. 

To lay a foundation for this and to provide a data-base for other researchers the Petros lab generated an epigenome atlas of neural progenitor cells of the mouse brain. This data includes scRNA-Seq, snATAC-Seq, CUT&Tag (H3K4me3, H3K27me3), CUT&RUN (H3K27ac), Hi-C and Capture-C. This data can be downloaded at the link below:  

https://www.nichd.nih.gov/research/atNICHD/Investigators/petros/data-sharing 

 

References

Datasets: https://www.nichd.nih.gov/research/atNICHD/Investigators/petros/data-sharing

Quattrocolo G, Fishell G, Petros TJ. Heterotopic Transplantations Reveal Environmental Influences on Interneuron Diversity and Maturation. Cell Rep. 2017 Oct 17;21(3):721-731. doi: 10.1016/j.celrep.2017.09.075. PMID: 29045839; PMCID: PMC5662128.

Dongjin R Lee, Christopher Rhodes, Apratim Mitra, Yajun Zhang, Dragan Maric, Ryan K Dale, Timothy J Petros (2022) Transcriptional heterogeneity of ventricular zone cells in the ganglionic eminences of the mouse forebrain eLife 11:e71864 https://doi.org/10.7554/eLife.71864

Rhodes, C. T., Thompson, J. J., Mitra, A., Asokumar, D., Lee, D. R., Lee, D. J., Zhang, Y., Jason, E., Dale, R. K., Rocha, P. P., & Petros, T. J. (2022). An epigenome atlas of neural progenitors within the embryonic mouse forebrain. Nature communications, 13(1), 4196. https://doi.org/10.1038/s41467-022-31793-4

 

Related Episodes

The Role of Histone Dopaminylation and Serotinylation in Neuronal Plasticity (Ian Maze)

Single-Cell Technologies using Microfluidics (Ben Hindson, CSO of 10x Genomics)

The Role of DNA Methylation in Epilepsy (Katja Kobow)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-08-25
Länk till avsnitt

Oncohistones as Drivers of Pediatric Brain Tumors (Nada Jabado)

In this episode of the Epigenetics Podcast, we caught up with Nada Jabado from McGill University to talk about her work on oncohistones as drivers of Pediatric Brain Tumors.

Nada Jabado and her team were amongst the first to identify mutations in Histone 3.3 Tails which lead to differentially remodeled chromatin in pediatric glioblastoma. Mutations that occur include the Lysine at position 27 and the Glycine at position 34. If those residues are mutated it will influence the equilibrium of chromatin associated proteins like the Polycomb Repressive Complex (PRC) and hence domains of heterochromatin will be shifted. This, in turn, will lead to differential gene expression and development of developmental disorders or cancer.

 

References

Schwartzentruber, J., Korshunov, A., Liu, X. Y., Jones, D. T., Pfaff, E., Jacob, K., Sturm, D., Fontebasso, A. M., Quang, D. A., Tönjes, M., Hovestadt, V., Albrecht, S., Kool, M., Nantel, A., Konermann, C., Lindroth, A., Jäger, N., Rausch, T., Ryzhova, M., Korbel, J. O., ? Jabado, N. (2012). Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature, 482(7384), 226?231. https://doi.org/10.1038/nature10833

Kleinman, C. L., Gerges, N., Papillon-Cavanagh, S., Sin-Chan, P., Pramatarova, A., Quang, D. A., Adoue, V., Busche, S., Caron, M., Djambazian, H., Bemmo, A., Fontebasso, A. M., Spence, T., Schwartzentruber, J., Albrecht, S., Hauser, P., Garami, M., Klekner, A., Bognar, L., Montes, J. L., ? Jabado, N. (2014). Fusion of TTYH1 with the C19MC microRNA cluster drives expression of a brain-specific DNMT3B isoform in the embryonal brain tumor ETMR. Nature genetics, 46(1), 39?44. https://doi.org/10.1038/ng.2849

Papillon-Cavanagh, S., Lu, C., Gayden, T., Mikael, L. G., Bechet, D., Karamboulas, C., Ailles, L., Karamchandani, J., Marchione, D. M., Garcia, B. A., Weinreb, I., Goldstein, D., Lewis, P. W., Dancu, O. M., Dhaliwal, S., Stecho, W., Howlett, C. J., Mymryk, J. S., Barrett, J. W., Nichols, A. C., ? Jabado, N. (2017). Impaired H3K36 methylation defines a subset of head and neck squamous cell carcinomas. Nature genetics, 49(2), 180?185. https://doi.org/10.1038/ng.3757

Chen, C., Deshmukh, S., Jessa, S., Hadjadj, D., Lisi, V., Andrade, A. F., Faury, D., Jawhar, W., Dali, R., Suzuki, H., Pathania, M., A, D., Dubois, F., Woodward, E., Hébert, S., Coutelier, M., Karamchandani, J., Albrecht, S., Brandner, S., De Jay, N., ? Jabado, N. (2020). Histone H3.3G34-Mutant Interneuron Progenitors Co-opt PDGFRA for Gliomagenesis. Cell, 183(6), 1617?1633.e22. https://doi.org/10.1016/j.cell.2020.11.012

Chaouch, A., Berlandi, J., Chen, C., Frey, F., Badini, S., Harutyunyan, A. S., Chen, X., Krug, B., Hébert, S., Jeibmann, A., Lu, C., Kleinman, C. L., Hasselblatt, M., Lasko, P., Shirinian, M., & Jabado, N. (2021). Histone H3.3 K27M and K36M mutations de-repress transposable elements through perturbation of antagonistic chromatin marks. Molecular cell, 81(23), 4876?4890.e7. https://doi.org/10.1016/j.molcel.2021.10.008

 

Related Episodes

Cancer and Epigenetics (David Jones)

Epigenetics & Glioblastoma: New Approaches to Treat Brain Cancer (Lucy Stead)

Targeting COMPASS to Cure Childhood Leukemia (Ali Shilatifard)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-08-11
Länk till avsnitt

Characterization of Epigenetic States in the Oligodendrocyte Lineage (Gonçalo Castelo-Branco)

In this episode of the Epigenetics Podcast, we caught up with Goncalo Castelo-Branco from the Karolinska Institute to talk about his work on the characterization of epigenetic states in the Oligodendrocyte Lineage.

The group from Gonçalo Castelo-Branco?s lab focuses on characterizing epigenetic states of oligodendrocytes, with the aim to understand their contribution to diseases like multiple sclerosis. To do this the group used single-cell RNA-Seq to identify sub-populations of oligodendrocytes. Furthermore, the team pioneered improvements in CUT&Tag and applied it to the single-cell space, as well as developing spatial CUT&Tag. More recently they used nanobodies in an optimised version of single cell CUT&Tag that allows simultaneous probing of three epigenomic modalities at single-cell resolution, using nanobody-Tn5 fusion proteins. The three modalities encompass chromatin accessibility as measured via ATAC-Seq and two histone post-transcriptional modifications.

 

References

Deng Y, Bartosovic M, Kukanja P, Zhang D, Liu Y, Su G, Enninful A, Bai Z, Castelo-Branco G, Fan R. Spatial-CUT&Tag: Spatially resolved chromatin modification profiling at the cellular level. Science. 2022 Feb 11;375(6581):681-686. doi: 10.1126/science.abg7216. Epub 2022 Feb 10. PMID: 35143307.

Winick-Ng W, Kukalev A, Harabula I, Zea-Redondo L, Szabó D, Meijer M, Serebreni L, Zhang Y, Bianco S, Chiariello AM, Irastorza-Azcarate I, Thieme CJ, Sparks TM, Carvalho S, Fiorillo L, Musella F, Irani E, Torlai Triglia E, Kolodziejczyk AA, Abentung A, Apostolova G, Paul EJ, Franke V, Kempfer R, Akalin A, Teichmann SA, Dechant G, Ungless MA, Nicodemi M, Welch L, Castelo-Branco G, Pombo A. Cell-type specialization is encoded by specific chromatin topologies. Nature. 2021 Nov;599(7886):684-691. doi: 10.1038/s41586-021-04081-2. Epub 2021 Nov 17. PMID: 34789882; PMCID: PMC8612935.

Bartosovic M, Kabbe M, Castelo-Branco G. Single-cell CUT&Tag profiles histone modifications and transcription factors in complex tissues. Nat Biotechnol. 2021 Jul;39(7):825-835. doi: 10.1038/s41587-021-00869-9. Epub 2021 Apr 12. PMID: 33846645; PMCID: PMC7611252.

Marek Bartosovic, Gonçalo Castelo-Branco. Multimodal chromatin profiling using nanobody-based single-cell CUT&Tag. bioRxiv. 2022.03.08.483459; doi: https://doi.org/10.1101/2022.03.08.483459

 

Related Episodes

Multiple challenges of CUT&Tag (Cassidee McDonough, Kyle Tanguay)

Chromatin Profiling: From ChIP to CUT&RUN, CUT&Tag and CUTAC (Steven Henikoff)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-07-28
Länk till avsnitt

Multiple challenges of ATAC-Seq, Points to Consider (Yuan Xue)

In this episode of the Epigenetics Podcast, we caught up with Active Motif?s own Yuan Xue to talk about some of the challenges of performing ATAC-Seq.

ATAC-Seq stands for Assay for Transposase-Accessible Chromatin with high-throughput sequencing and was initially described by Jason Buenrostro in 2013. The ATAC-Seq method relies on next-generation sequencing (NGS) library construction using the hyperactive transposase Tn5. NGS adapters are loaded onto the transposase, which allows simultaneous fragmentation of chromatin and integration of those adapters into open chromatin regions. ATAC-Seq is an attractive method to start your epigenetic journey. Whether you want to analyze the state of the chromatin in your sample or compare the chromatin state before and after a special treatment, ATAC-Seq allows you to investigate genome-wide chromatin changes and can offer guidelines about which epigenetic modification or transcription factor should be studied next in the follow-up experiments and which method should be used to study them.

In this Episode we go through the Protocol in detail and discuss potential challenges and points to pay attention to when starting your first ATAC-Seq experiment.

 

References

ATAC-Seq Resource Center

Complete Guide to Understanding and Using ATAC-Seq

Beginner?s Guide to Understanding Single-Cell ATAC-Seq

Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y., & Greenleaf, W. J. (2013). Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nature methods, 10(12), 1213?1218. https://doi.org/10.1038/nmeth.2688

Buenrostro, J. D., Wu, B., Litzenburger, U. M., Ruff, D., Gonzales, M. L., Snyder, M. P., Chang, H. Y., & Greenleaf, W. J. (2015). Single-cell chromatin accessibility reveals principles of regulatory variation. Nature, 523(7561), 486?490. https://doi.org/10.1038/nature14590

Cusanovich, D. A., Daza, R., Adey, A., Pliner, H. A., Christiansen, L., Gunderson, K. L., Steemers, F. J., Trapnell, C., & Shendure, J. (2015). Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science (New York, N.Y.), 348(6237), 910?914. https://doi.org/10.1126/science.aab1601

Podcast: ATAC-Seq, scATAC-Seq and Chromatin Dynamics in Single-Cells (Jason Buenrostro)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-07-14
Länk till avsnitt

The Effect of lncRNAs on Chromatin and Gene Regulation (John Rinn)

In this episode of the Epigenetics Podcast, we caught up with John Rinn from the University of Colorado in Boulder to talk about his work on the role of lncRNAs in gene expression and nuclear organization.

The Rinn Lab pioneered the approach of screening the human genome for long noncoding RNAs (lncRNAs). More recently, the lab has shifted focus from measuring the number of lncRNAs to finding lncRNAs that have a distinct biological function in human health and disease. One example of such a lncRNA is FIRRE, which is present in all animals, however the sequence is not conserved, except for in primates. FIRRE contains many interesting features, such as repeat sequences and CTCF binding sites. In absence of FIRRE, defects in the immune system can be observed and also some brain defects may also be observed.

 

References

Carter, T., Singh, M., Dumbovic, G., Chobirko, J. D., Rinn, J. L., & Feschotte, C. (2022). Mosaic cis-regulatory evolution drives transcriptional partitioning of HERVH endogenous retrovirus in the human embryo. eLife, 11, e76257. Advance online publication. https://doi.org/10.7554/eLife.76257

Long, Y., Hwang, T., Gooding, A. R., Goodrich, K. J., Rinn, J. L., & Cech, T. R. (2020). RNA is essential for PRC2 chromatin occupancy and function in human pluripotent stem cells. Nature Genetics, 52(9), 931?938. https://doi.org/10.1038/s41588-020-0662-x

Kelley, D., & Rinn, J. (2012). Transposable elements reveal a stem cell-specific class of long noncoding RNAs. Genome biology, 13(11), R107. https://doi.org/10.1186/gb-2012-13-11-r107

Khalil, A. M., Guttman, M., Huarte, M., Garber, M., Raj, A., Rivea Morales, D., Thomas, K., Presser, A., Bernstein, B. E., van Oudenaarden, A., Regev, A., Lander, E. S., & Rinn, J. L. (2009). Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proceedings of the National Academy of Sciences, 106(28), 11667?11672. https://doi.org/10.1073/pnas.0904715106

Guttman, M., Amit, I., Garber, M., French, C., Lin, M. F., Feldser, D., Huarte, M., Zuk, O., Carey, B. W., Cassady, J. P., Cabili, M. N., Jaenisch, R., Mikkelsen, T. S., Jacks, T., Hacohen, N., Bernstein, B. E., Kellis, M., Regev, A., Rinn, J. L., & Lander, E. S. (2009). Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature, 458(7235), 223?227. https://doi.org/10.1038/nature07672

 

Related Episodes

The Role of lncRNAs in Tumor Growth and Treatment (Sarah Diermeier)

The Role of Small RNAs in Transgenerational Inheritance in C. elegans (Oded Rechavi)

Chromatin Structure and Dynamics at Ribosomal RNA Genes (Tom Moss)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-06-30
Länk till avsnitt

Epigenetic Clocks and Biomarkers of Ageing (Morgan Levine)

 In this episode of the Epigenetics Podcast, we caught up with Morgan Levine from Altos lab to talk about her work on Epigenetic Clocks and Biomarkers of Ageing.

The Levine Lab focuses on deciphering mechanisms that lead to epigenetic ageing, which can be measured by epigenetic clocks. Epigenetic clocks were first described in 2011 by Bocklandt et al.. Later-on, the Horvath and the Hannum clock were described by using a combination of CpGs to calculate biological/epigenetic age in contrast to chronological age.

The Levine Lab themselves worked on generating an advanced version of an Epigenetic clock, called "DNAm PhenoAge" that will now be used, and not only in human samples. The team now moves to mouse models and to cells in a dish and using those models to investigate the mechanisms behind epigenetic aging.

 

References

Liu, Z., Leung, D., Thrush, K., Zhao, W., Ratliff, S., Tanaka, T., Schmitz, L. L., Smith, J. A., Ferrucci, L., & Levine, M. E. (2020). Underlying features of epigenetic aging clocks in vivo and in vitro. Aging cell, 19(10), e13229. https://doi.org/10.1111/acel.13229

Levine, M. E., Lu, A. T., Quach, A., Chen, B. H., Assimes, T. L., Bandinelli, S., Hou, L., Baccarelli, A. A., Stewart, J. D., Li, Y., Whitsel, E. A., Wilson, J. G., Reiner, A. P., Aviv, A., Lohman, K., Liu, Y., Ferrucci, L., & Horvath, S. (2018). An epigenetic biomarker of aging for lifespan and healthspan. Aging, 10(4), 573?591. https://doi.org/10.18632/aging.101414

Levine, M., McDevitt, R. A., Meer, M., Perdue, K., Di Francesco, A., Meade, T., Farrell, C., Thrush, K., Wang, M., Dunn, C., Pellegrini, M., de Cabo, R., & Ferrucci, L. (2020). A rat epigenetic clock recapitulates phenotypic aging and co-localizes with heterochromatin. eLife, 9, e59201. https://doi.org/10.7554/eLife.59201

Kuo, C. L., Pilling, L. C., Atkins, J. C., Masoli, J., Delgado, J., Tignanelli, C., Kuchel, G., Melzer, D., Beckman, K. B., & Levine, M. (2020). COVID-19 severity is predicted by earlier evidence of accelerated aging. medRxiv : the preprint server for health sciences, 2020.07.10.20147777. https://doi.org/10.1101/2020.07.10.20147777

 

Related Episodes

Aging and Epigenetics (Peter Tessarz)

Epigenetic Mechanisms of Aging and Longevity (Shelley Berger)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-06-23
Länk till avsnitt

Epigenetic and Metabolic Regulation of Early Development (Jan ?ylicz)

In this episode of the Epigenetics Podcast, we caught up with Jan ?ylicz from the Novo Nordisk Foundation Center for Stem Cell Medicine to talk about his work on epigenetic and metabolic regulation of early development.

The focus of the ?ylicz Lab is studying early development and how this process is influenced by epigenetic factors. In more detail, the Team focuses on the function of chromatin modifiers in this process. Primed pluripotent epiblasts in vivo show a distinct chromatin landscape that is characterized by high levels of histone H3 lysine 9 dimethylation (H3K9me2) and rearranged Polycomb-associated histone H3 lysine 27 trimethylation (H3K27me3) at thousands of genes along the genome. However, the function of only about 100 loci is impaired. The ?ylicz Lab tries to understand this process behind and also the cause of this discrepancy.

 

References

?ylicz, J. J., Bousard, A., ?umer, K., Dossin, F., Mohammad, E., da Rocha, S. T., Schwalb, B., Syx, L., Dingli, F., Loew, D., Cramer, P., & Heard, E. (2019). The Implication of Early Chromatin Changes in X Chromosome Inactivation. Cell, 176(1?2), 182-197.e23. https://doi.org/10.1016/j.cell.2018.11.041

Dossin, F., Pinheiro, I., ?ylicz, J. J., Roensch, J., Collombet, S., Le Saux, A., Chelmicki, T., Attia, M., Kapoor, V., Zhan, Y., Dingli, F., Loew, D., Mercher, T., Dekker, J., & Heard, E. (2020). SPEN integrates transcriptional and epigenetic control of X-inactivation. Nature, 578(7795), 455?460. https://doi.org/10.1038/s41586-020-1974-9

 

Related Episodes

Epigenetics and X-Inactivation (Edith Heard)

The Effects of Early Life Stress on Mammalian Development (Catherine J. Peña)

DNA Methylation and Mammalian Development (Déborah Bourc'his)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-06-09
Länk till avsnitt

Multiple challenges of CUT&Tag (Cassidee McDonough, Kyle Tanguay)

In this episode of the Epigenetics Podcast, we caught up with Active Motif scientists Casidee McDonough from Epigenetic Services and Kyle Tanguay from R&D to talk about technical details of the CUT&Tag protocol and current developments around this method in our R&D Team. 

CUT&Tag, which is short for Cleavage Under Targets and Tagmentation, is a molecular biology method that is used to investigate interactions between proteins and DNA and to identify DNA binding sites for their protein of interest. Although CUT&Tag is similar in some ways to ChIP assays, the starting material for CUT&Tag is live, permeabilized cells or isolated cell nuclei, rather than cells or tissue that have been crosslinked with formaldehyde as is the case when performing ChIP. The CUT&Tag method is very sensitive and has been reported to work with as few as 60 cells for some histone modifications. The ability to work with such small numbers of cells is an advantage for researchers working on specific cell types, such as rare neuronal populations, pancreatic islets, or stem cells that are difficult to obtain in large numbers. 

In this Episode we discuss the CUT&Tag workflow in detail, talk about the challenges and pitfalls, give guidelines on how to do a good CUT&Tag experiment and offer a glimpse into the future of CUT&Tag product development at Active Motif. 

 

References

Comprehensive Guide to Understanding and Using CUT&Tag Assays

Library QC for ATAC-Seq and CUT&Tag | AKA ?Does My Library Look Okay??

Kaya-Okur, H.S., Wu, S.J., Codomo, C.A. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat Commun 10, 1930 (2019). https://doi.org/10.1038/s41467-019-09982-5

Podcast: Chromatin Profiling: From ChIP to CUT&RUN, CUT&Tag and CUTAC (Steven Henikoff)

CUT&Tag-validated antibodies

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-05-26
Länk till avsnitt

The Role of Histone Dopaminylation and Serotinylation in Neuronal Plasticity (Ian Maze)

In this episode of the Epigenetics Podcast, we caught up with Ian Maze from Ichan School of Medicine at Mount Sinai and a Howard Hughes Medical Institute (HHMI) Investigator to talk about his work on the role of histone dopaminylation and serotinylation in neuronal plasticity.

The Maze group focuses on understanding the complex interplay between chromatin regulatory mechanisms in brain and neuronal plasticity. The lab places an emphasis on psychiatric disorders associated with monoaminergic (e.g., serotonin, dopamine, etc.) dysfunction, such as major depressive disorder and drug addiction. In particular the Maze team has investigated cocaine addiction and its effect on chromatin by serotonylation and dopaminylation of Histone H3 Tails.

 

References

Maze, I., Covington, H. E., Dietz, D. M., LaPlant, Q., Renthal, W., Russo, S. J., Mechanic, M., Mouzon, E., Neve, R. L., Haggarty, S. J., Ren, Y., Sampath, S. C., Hurd, Y. L., Greengard, P., Tarakhovsky, A., Schaefer, A., & Nestler, E. J. (2010). Essential Role of the Histone Methyltransferase G9a in Cocaine-Induced Plasticity. Science, 327(5962), 213?216. https://doi.org/10.1126/science.1179438

Farrelly, L. A., Thompson, R. E., Zhao, S., Lepack, A. E., Lyu, Y., Bhanu, N. V., Zhang, B., Loh, Y.-H. E., Ramakrishnan, A., Vadodaria, K. C., Heard, K. J., Erikson, G., Nakadai, T., Bastle, R. M., Lukasak, B. J., Zebroski, H., Alenina, N., Bader, M., Berton, O., ? Maze, I. (2019). Histone serotonylation is a permissive modification that enhances TFIID binding to H3K4me3. Nature, 567(7749), 535?539. https://doi.org/10.1038/s41586-019-1024-7

Lepack, A. E., Werner, C. T., Stewart, A. F., Fulton, S. L., Zhong, P., Farrelly, L. A., Smith, A. C. W., Ramakrishnan, A., Lyu, Y., Bastle, R. M., Martin, J. A., Mitra, S., O?Connor, R. M., Wang, Z.-J., Molina, H., Turecki, G., Shen, L., Yan, Z., Calipari, E. S., ? Maze, I. (2020). Dopaminylation of histone H3 in ventral tegmental area regulates cocaine seeking. Science, 368(6487), 197?201. https://doi.org/10.1126/science.aaw8806

 

Related Episodes

Development of Integrative Machine Learning Tools for Neurodegenerative Diseases (Enrico Glaab)

Epigenetic Influence on Memory Formation and Inheritance (Isabelle Mansuy)

CpG Islands, DNA Methylation, and Disease (Sir Adrian Bird)

 

Contact

Active Motif on Twitter

Epigenetics Podcast on Twitter

Active Motif on LinkedIn

Active Motif on Facebook

Email: [email protected]

2022-05-12
Länk till avsnitt
Hur lyssnar man på podcast?

En liten tjänst av I'm With Friends. Finns även på engelska.
Uppdateras med hjälp från iTunes.