scispace - formally typeset
Search or ask a question
Author

Tamara J. McLennan

Bio: Tamara J. McLennan is an academic researcher from Walter and Eliza Hall Institute of Medical Research. The author has contributed to research in topics: Germline & Somatic hypermutation. The author has an hindex of 5, co-authored 6 publications receiving 208 citations.

Papers
More filters
Journal ArticleDOI
TL;DR: This article showed that lack of the histone acetyltransferase MOZ (MYST3/KAT6A) phenocopies DiGeorge syndrome, and the MOZ complex occupies the Tbx1 locus, promoting its expression and histone 3lysine 9 acetylation.

77 citations

01 Jan 2012
TL;DR: It is shown that lack of the histone acetyltransferase MOZ phenocopies DiGeorge syndrome, and the MOZ complex occupies the Tbx1 locus, promoting its expression and histone 3 lysine 9 acetylation.
Abstract: SUMMARY DiGeorge syndrome, caused by a 22q11 microdeletion or mutation of the TBX1 gene, varies in severity greatly, even among monozygotic twins. Epigenetic phenomena have been invoked to explain phenotypic differences in individuals of identical genetic composition, although specific chromatin modifications relevant to DiGeorge syndrome are elusive. Here we show that lack of the histone acetyltransferase MOZ (MYST3/KAT6A) phenocopies DiGeorge syndrome, and the MOZ complex occupies the Tbx1 locus, promoting its expression and histone 3 lysine 9 acetylation. Importantly, DiGeorge syndrome-like anomalies are present in mice with homozygous mutation of Moz and in heterozygous Moz mutants when combined with Tbx1 haploinsufficiency or oversupply of retinoic acid. Conversely, a Tbx1 transgene rescues the heart phenotype in Moz mutants. Our data reveal a molecular mechanism for a specific chromatin modification of the Tbx1 locus intersecting with an environmental determinant, modeling variability in DiGeorge syndrome.

75 citations

Book ChapterDOI
TL;DR: Using ChIP on mouse embryos, the presence or absence of specific proteins and chromatin modifications at genomic loci in vivo during mammalian development is documented.
Abstract: During prenatal development, a large number of different cell types are formed, the vast majority of which contain identical genetic material. The basis of the great variety in cell phenotype and function is the differential expression of the approximately 25,000 genes in the mammalian genome. Transcriptional activity is regulated at many levels by proteins, including members of the basal transcriptional apparatus, DNA-binding transcription factors, and chromatin-binding proteins. Importantly, chromatin structure dictates the availability of a specific genomic locus for transcriptional activation as well as the efficiency, with which transcription can occur. Chromatin immunoprecipitation (ChIP) is a method to assess if chromatin modifications or proteins are present at a specific locus. ChIP involves the cross linking of DNA and associated proteins and immunoprecipitation using specific antibodies to DNA-associated proteins followed by examination of the co-precipitated DNA sequences or proteins. In the last few years, ChIP has become an essential technique for scientists studying transcriptional regulation and chromatin structure. Using ChIP on mouse embryos, we can document the presence or absence of specific proteins and chromatin modifications at genomic loci in vivo during mammalian development. Here, we describe a ChIP technique adapted for mouse embryos.

16 citations

Journal ArticleDOI
TL;DR: It is hypothesize that, by terminating reelin signaling, SOCS6 and SOCS7 may allow new cycles of reelin signaled to occur and that these may be essential for cortical neuron migration.
Abstract: Mutations of the reelin gene cause severe defects in cerebral cortex development and profound intellectual impairment. While many aspects of the reelin signaling pathway have been identified, the molecular and ultimate cellular consequences of reelin signaling remain unknown. Specifically, it is unclear if termination of reelin signaling is as important for normal cortical neuron migration as activation of reelin signaling. Using mice that are single or double deficient, we discovered that combined loss of the suppressors of cytokine signaling, SOCS6 and SOCS7, recapitulated the cortical layer inversion seen in mice lacking reelin and led to a dramatic increase in the reelin signaling molecule disabled (DAB1) in the cortex. The SRC homology domains of SOCS6 and SOCS7 bound DAB1 ex vivo. Mutation of DAB1 greatly diminished binding and protected from degradation by SOCS6. Phosphorylated DAB1 was elevated in cortical neurons in the absence of SOCS6 and SOCS7. Thus, constitutive activation of reelin signaling was observed to be equally detrimental as lack of activation. We hypothesize that, by terminating reelin signaling, SOCS6 and SOCS7 may allow new cycles of reelin signaling to occur and that these may be essential for cortical neuron migration.

16 citations


Cited by
More filters
Journal ArticleDOI
01 Nov 2019-Science
TL;DR: This process of “clonal hematopoiesis,” including the mechanisms by which it arises and the current state of knowledge regarding its effects on human health is reviewed, including the prevalence and clinical associations of somatic, clonal mutations in blood cells of individuals without hematologic malignancies.
Abstract: As people age, their tissues accumulate an increasing number of somatic mutations. Although most of these mutations are of little or no functional consequence, a mutation may arise that confers a fitness advantage on a cell. When this process happens in the hematopoietic system, a substantial proportion of circulating blood cells may derive from a single mutated stem cell. This outgrowth, called "clonal hematopoiesis," is highly prevalent in the elderly population. Here we discuss recent advances in our knowledge of clonal hematopoiesis, its relationship to malignancies, its link to nonmalignant diseases of aging, and its potential impact on immune function. Clonal hematopoiesis provides a glimpse into the process of mutation and selection that likely occurs in all somatic tissues.

504 citations

Journal ArticleDOI
29 Jun 2022-Blood
TL;DR: The authors, a group with expertise in the clinical, pathologic and genetic aspects of these disorders, developed the International Consensus Classification (ICC), aimed at facilitating diagnosis and prognostication of these neoplasms, improving treatment of affected patients, and allowing the design of innovative clinical trials.

444 citations

Journal ArticleDOI
TL;DR: Special focus will be given to epigenetic regulators that drive the commitment of cardiomyogenic cells from nascent mesoderm and their differentiation into chamber-specific myocytes, as well as regulation of myocardial trabeculation.
Abstract: The heart is the first organ to form during embryonic development. Given the complex nature of cardiac differentiation and morphogenesis, it is not surprising that some form of congenital heart disease is present in ≈1 percent of newborns. The molecular determinants of heart development have received much attention over the past several decades. This has been driven in large part by an interest in understanding the causes of congenital heart disease coupled with the potential of using knowledge from developmental biology to generate functional cells and tissues that could be used for regenerative medicine purposes. In this review, we highlight the critical signaling pathways and transcription factor networks that regulate cardiomyocyte lineage specification in both in vivo and in vitro models. Special focus will be given to epigenetic regulators that drive the commitment of cardiomyogenic cells from nascent mesoderm and their differentiation into chamber-specific myocytes, as well as regulation of myocardial trabeculation.

160 citations

Journal ArticleDOI
Jonathan B. Baell1, Jonathan B. Baell2, David J. Leaver2, S.J. Hermans3, Gemma L. Kelly4, Gemma L. Kelly5, Margs S. Brennan5, Margs S. Brennan4, Natalie L. Downer5, Nghi H. Nguyen2, Johannes Wichmann5, Johannes Wichmann4, Helen M. McRae5, Helen M. McRae4, Yuquing Yang4, Yuquing Yang5, Benjamin Cleary2, H. Rachel Lagiakos2, Stephen Mieruszynski5, Stephen Mieruszynski4, Guido Pacini5, Hannah K. Vanyai5, Hannah K. Vanyai4, Maria I. Bergamasco5, Maria I. Bergamasco4, Rose E. May5, Bethany K. Davey5, Kimberly J. Morgan4, Kimberly J. Morgan5, Andrew J. Sealey4, Andrew J. Sealey5, Beinan Wang5, Beinan Wang4, Beinan Wang6, Natasha Zamudio5, Natasha Zamudio4, Stephen Wilcox5, Stephen Wilcox4, Alexandra L. Garnham4, Alexandra L. Garnham5, Bilal N. Sheikh4, Bilal N. Sheikh5, Brandon J. Aubrey5, Brandon J. Aubrey4, Karen Doggett4, Karen Doggett5, M.C. Chung3, Melanie de Silva5, John D. Bentley7, Pat Pilling7, Meghan Hattarki7, Olan Dolezal7, Matthew L. Dennis7, Hendrik Falk4, Hendrik Falk5, Bin Ren7, Susan A. Charman2, Karen L. White2, Jai Rautela4, Jai Rautela5, Andrea Newbold8, Edwin D. Hawkins5, Edwin D. Hawkins4, Ricky W. Johnstone8, Nicholas D. Huntington4, Nicholas D. Huntington5, Thomas S. Peat7, Joan K. Heath5, Joan K. Heath4, Andreas Strasser4, Andreas Strasser5, Michael W. Parker3, Michael W. Parker4, Gordon K. Smyth4, Gordon K. Smyth5, Ian P. Street5, Ian P. Street4, Brendon J. Monahan5, Brendon J. Monahan4, Anne K. Voss4, Anne K. Voss5, Tim Thomas5, Tim Thomas4 
01 Aug 2018-Nature
TL;DR: Biochemical and structural studies demonstrate that these compounds are reversible competitors of acetyl coenzyme A and inhibit MYST-catalysed histone acetylation, induce cell cycle exit and cellular senescence without causing DNA damage, and arrest lymphoma progression in mouse models.
Abstract: Acetylation of histones by lysine acetyltransferases (KATs) is essential for chromatin organization and function1. Among the genes coding for the MYST family of KATs (KAT5-KAT8) are the oncogenes KAT6A (also known as MOZ) and KAT6B (also known as MORF and QKF)2,3. KAT6A has essential roles in normal haematopoietic stem cells4-6 and is the target of recurrent chromosomal translocations, causing acute myeloid leukaemia7,8. Similarly, chromosomal translocations in KAT6B have been identified in diverse cancers8. KAT6A suppresses cellular senescence through the regulation of suppressors of the CDKN2A locus9,10, a function that requires its KAT activity10. Loss of one allele of KAT6A extends the median survival of mice with MYC-induced lymphoma from 105 to 413 days11. These findings suggest that inhibition of KAT6A and KAT6B may provide a therapeutic benefit in cancer. Here we present highly potent, selective inhibitors of KAT6A and KAT6B, denoted WM-8014 and WM-1119. Biochemical and structural studies demonstrate that these compounds are reversible competitors of acetyl coenzyme A and inhibit MYST-catalysed histone acetylation. WM-8014 and WM-1119 induce cell cycle exit and cellular senescence without causing DNA damage. Senescence is INK4A/ARF-dependent and is accompanied by changes in gene expression that are typical of loss of KAT6A function. WM-8014 potentiates oncogene-induced senescence in vitro and in a zebrafish model of hepatocellular carcinoma. WM-1119, which has increased bioavailability, arrests the progression of lymphoma in mice. We anticipate that this class of inhibitors will help to accelerate the development of therapeutics that target gene transcription regulated by histone acetylation.

159 citations

Journal ArticleDOI
17 May 2018-Cell
TL;DR: It is provided evidence that necroptosis, pyroPTosis, or autophagy does not substantially substitute for the loss of apoptosis and suggests that morphogenesis can proceed entirely without apoptosis mediated by these proteins and possibly without cell death in general.

144 citations