101
|
Oulhen N, Wessel GM. Differential Nanos 2 protein stability results in selective germ cell accumulation in the sea urchin. Dev Biol 2016; 418:146-156. [PMID: 27424271 DOI: 10.1016/j.ydbio.2016.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 06/21/2016] [Accepted: 07/12/2016] [Indexed: 01/18/2023]
Abstract
Nanos is a translational regulator required for the survival and maintenance of primordial germ cells. In the sea urchin, Strongylocentrotus purpuratus (Sp), Nanos 2 mRNA is broadly transcribed but accumulates specifically in the small micromere (sMic) lineage, in part because of the 3'UTR element GNARLE leads to turnover in somatic cells but retention in the sMics. Here we found that the Nanos 2 protein is also selectively stabilized; it is initially translated throughout the embryo but turned over in the future somatic cells and retained only in the sMics, the future germ line in this animal. This differential stability of Nanos protein is dependent on the open reading frame (ORF), and is independent of the sumoylation and ubiquitylation pathways. Manipulation of the ORF indicates that 68 amino acids in the N terminus of the Nanos protein are essential for its stability in the sMics whereas a 45 amino acid element adjacent to the zinc fingers targets its degradation. Further, this regulation of Nanos protein is cell autonomous, following formation of the germ line. These results are paradigmatic for the unique presence of Nanos in the germ line by a combination of selective RNA retention, distinctive translational control mechanisms (Oulhen et al., 2013), and now also by defined Nanos protein stability.
Collapse
Affiliation(s)
- Nathalie Oulhen
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting St., Providence, RI 02912, USA
| | - Gary M Wessel
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting St., Providence, RI 02912, USA.
| |
Collapse
|
102
|
Poon J, Wessel GM, Yajima M. An unregulated regulator: Vasa expression in the development of somatic cells and in tumorigenesis. Dev Biol 2016; 415:24-32. [PMID: 27179696 PMCID: PMC4902722 DOI: 10.1016/j.ydbio.2016.05.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 02/08/2023]
Abstract
Growing evidence in diverse organisms shows that genes originally thought to function uniquely in the germ line may also function in somatic cells, and in some cases even contribute to tumorigenesis. Here we review the somatic functions of Vasa, one of the most conserved "germ line" factors among metazoans. Vasa expression in somatic cells is tightly regulated and often transient during normal development, and appears to play essential roles in regulation of embryonic cells and regenerative tissues. Its dysregulation, however, is believed to be an important element of tumorigenic cell regulation. In this perspectives paper, we propose how some conserved functions of Vasa may be selected for somatic cell regulation, including its potential impact on efficient and localized translational activities and in some cases on cellular malfunctioning and tumorigenesis.
Collapse
Affiliation(s)
- Jessica Poon
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA
| | - Gary M Wessel
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA
| | - Mamiko Yajima
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA.
| |
Collapse
|
103
|
Bryant SA, Herdy JR, Amemiya CT, Smith JJ. Characterization of Somatically-Eliminated Genes During Development of the Sea Lamprey (Petromyzon marinus). Mol Biol Evol 2016; 33:2337-44. [PMID: 27288344 DOI: 10.1093/molbev/msw104] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The sea lamprey (Petromyzon marinus) is a basal vertebrate that undergoes developmentally programmed genome rearrangements (PGRs) during early development. These events facilitate the elimination of ∼20% of the genome from the somatic cell lineage, resulting in distinct somatic and germline genomes. Thus far only a handful of germline-specific genes have been definitively identified within the estimated 500 Mb of DNA that is deleted during PGR, although a few thousand germline-specific genes are thought to exist. To improve our understanding of the evolutionary/developmental logic of PGR, we generated computational predictions to identify candidate germline-specific genes within a new transcriptomic dataset derived from adult germline and the early embryonic stages during which PGR occurs. Follow-up validation studies identified 44 germline-specific genes and further characterized patterns of transcription and DNA loss during early embryogenesis. Expression analyses reveal that many of these genes are differentially expressed during early embryogenesis and presumably function in the early development of the germline. Ontology analyses indicate that many of these germline-specific genes play known roles in germline development, pluripotency, and oncogenesis (when misexpressed). These studies provide support for the theory that PGR serves to segregate molecular functions related to germline development/pluripotency in order to prevent their potential misexpression in somatic cells. This larger set of eliminated genes also allows us to extend the evolutionary/developmental breadth of this theory, as some deleted genes (or their gnathostome homologs) appear to be associated with the early development of somatic lineages, perhaps through the evolution of novel functions within gnathostome lineages.
Collapse
Affiliation(s)
| | | | - Chris T Amemiya
- Benaroya Research Institute at Virginia Mason, Seattle Department of Biology, University of Washington, Seattle
| | | |
Collapse
|
104
|
Abstract
Epigenetic regulation of chromatin structure is a fundamental process for eukaryotes. Regulators include DNA methylation, microRNAs and chromatin modifications. Within the chromatin modifiers, one class of enzymes that can functionally bind and modify chromatin, through the removal of methyl marks, is the histone lysine demethylases. Here, we summarize the current findings of the 13 known histone lysine demethylases in Drosophila melanogaster, and discuss the critical role of these histone-modifying enzymes in the maintenance of genomic functions. Additionally, as histone demethylase dysregulation has been identified in cancer, we discuss the advantages for using Drosophila as a model system to study tumorigenesis.
Collapse
Affiliation(s)
- Andreana Holowatyj
- a Department of Oncology ; Wayne State University School of Medicine ; Detroit , MI USA
| | | | | |
Collapse
|
105
|
A six gene expression signature defines aggressive subtypes and predicts outcome in childhood and adult acute lymphoblastic leukemia. Oncotarget 2016; 6:16527-42. [PMID: 26001296 PMCID: PMC4599287 DOI: 10.18632/oncotarget.4113] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 04/22/2015] [Indexed: 11/25/2022] Open
Abstract
Abnormal gene expression in cancer represents an under-explored source of cancer markers and therapeutic targets. In order to identify gene expression signatures associated with survival in acute lymphoblastic leukemia (ALL), a strategy was designed to search for aberrant gene activity, which consists of applying several filters to transcriptomic datasets from two pediatric ALL studies. Six genes whose expression in leukemic blasts was associated with prognosis were identified:three genes predicting poor prognosis (AK022211, FASTKD1 and STARD4) and three genes associated with a favorable outcome (CAMSAP1, PCGF6 and SH3RF3). Combining the expression of these 6 genes could successfully predict prognosis not only in the two discovery pediatric ALL studies, but also in two independent validation cohorts of adult patients, one from a publicly available study and one consisting of 62 newly recruited Chinese patients. Moreover, our data demonstrate that our six gene based test is particularly efficient in stratifying MLL or BCR.ABL negative patients. Finally, common biological traits characterizing aggressive forms of ALL in both children and adults were found, including features of dormant hematopoietic stem cells, suggesting new therapeutic strategies.
Collapse
|
106
|
Christofi T, Apidianakis Y. Drosophila and the hallmarks of cancer. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2016; 135:79-110. [PMID: 23615878 DOI: 10.1007/10_2013_190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
: Cancer was the disease of the twentieth century. Today it is still a leading cause of death worldwide despite being intensively investigated. Abundant knowledge exists regarding the pathological and molecular mechanisms that drive healthy cells to become malignant and form metastatic tumors. The relation of oncogenes and tumor suppressors to the genetic trigger of carcinogenesis is unquestionable. However, the development of the disease requires many characteristics that due to their proven role in cancer are collectively described as the "hallmarks of cancer." We highlight here the historic discoveries made using the model organism Drosophila melanogaster and its contributions to biomedical and cancer research. Flies are utilized as a model organism for the investigation of each and every aspect of cancer hallmarks. Due to the significant conservation between flies and mammals at the signaling and tissue physiology level it is possible to explore the genes and mechanisms responsible for cancer pathogenesis in flies. Recent Drosophila studies suggest novel aspects of therapeutic intervention and are expected to guide cancer research in the twenty-first century.
Collapse
|
107
|
Hickman ES, Lomax ME, Jakobsen BK. Antigen Selection for Enhanced Affinity T-Cell Receptor-Based Cancer Therapies. ACTA ACUST UNITED AC 2016; 21:769-85. [PMID: 26993321 DOI: 10.1177/1087057116637837] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/15/2016] [Indexed: 12/11/2022]
Abstract
Evidence of adaptive immune responses in the prevention of cancer has been accumulating for decades. Spontaneous T-cell responses occur in multiple indications, bringing the study of de novo expressed cancer antigens to the fore and highlighting their potential as targets for cancer immunotherapy. Circumventing the immune-suppressive mechanisms that maintain tumor tolerance and driving an antitumor cytotoxic T-cell response in cancer patients may eradicate the tumor or block disease progression. Multiple strategies are being pursued to harness the cytotoxic potential of T cells clinically. Highly promising results are now emerging. The focus of this review is the target discovery process for cancer immune therapeutics based on affinity-matured T-cell receptors (TCRs). Target cancer antigens in the context of adoptive cell transfer technologies and soluble biologic agents are discussed. To appreciate the impact of TCR-based technology and understand the TCR discovery process, it is necessary to understand key differences between TCR-based therapy and other immunotherapy approaches. The review first summarizes key advances in the cancer immunotherapy field and then discusses the opportunities that TCR technology provides. The nature and breadth of molecular targets that are tractable to this approach are discussed, together with the challenges associated with finding them.
Collapse
|
108
|
Reevaluation of whether a soma-to-germ-line transformation extends lifespan in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2016; 113:3591-6. [PMID: 26976573 DOI: 10.1073/pnas.1523402113] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The germ lineage is considered to be immortal. In the quest to extend lifespan, a possible strategy is to drive germ-line traits in somatic cells, to try to confer some of the germ lineage's immortality on the somatic body. Notably, a study in Caenorhabditis elegans suggested that expression of germ-line genes in the somatic cells of long-lived daf-2 mutants confers some of daf-2's long lifespan. Specifically, mRNAs encoding components of C. elegans germ granules (P granules) were up-regulated in daf-2 mutant worms, and knockdown of individual P-granule and other germ-line genes in daf-2 young adults modestly reduced their lifespan. We investigated the contribution of a germ-line program to daf-2's long lifespan and also tested whether other mutants known to express germ-line genes in their somatic cells are long-lived. Our key findings are as follows. (i) We could not detect P-granule proteins in the somatic cells of daf-2 mutants by immunostaining or by expression of a P-granule transgene. (ii) Whole-genome transcript profiling of animals lacking a germ line revealed that germ-line transcripts are not up-regulated in the soma of daf-2 worms compared with the soma of control worms. (iii) Simultaneous removal of multiple P-granule proteins or the entire germ-line program from daf-2 worms did not reduce their lifespan. (iv) Several mutants that robustly express a broad spectrum of germ-line genes in their somatic cells are not long-lived. Together, our findings argue against the hypothesis that acquisition of a germ-cell program in somatic cells increases lifespan and contributes to daf-2's long lifespan.
Collapse
|
109
|
Abstract
The ciliate Oxytricha is a microbial eukaryote with two genomes, one of which experiences extensive genome remodeling during development. Each round of conjugation initiates a cascade of events that construct a transcriptionally active somatic genome from a scrambled germline genome, with considerable help from both long and small noncoding RNAs. This process of genome remodeling entails massive DNA deletion and reshuffling of remaining DNA segments to form functional genes from their interrupted and scrambled germline precursors. The use of Oxytricha as a model system provides an opportunity to study an exaggerated form of programmed genome rearrangement. Furthermore, studying the mechanisms that maintain nuclear dimorphism and mediate genome rearrangement has demonstrated a surprising plasticity and diversity of noncoding RNA pathways, with new roles that go beyond conventional gene silencing. Another aspect of ciliate genetics is their unorthodox patterns of RNA-mediated, epigenetic inheritance that rival Mendelian inheritance. This review takes the reader through the key experiments in a model eukaryote that led to fundamental discoveries in RNA biology and pushes the biological limits of DNA processing.
Collapse
|
110
|
Ku HY, Gangaraju VK, Qi H, Liu N, Lin H. Tudor-SN Interacts with Piwi Antagonistically in Regulating Spermatogenesis but Synergistically in Silencing Transposons in Drosophila. PLoS Genet 2016; 12:e1005813. [PMID: 26808625 PMCID: PMC4726654 DOI: 10.1371/journal.pgen.1005813] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 12/23/2015] [Indexed: 11/23/2022] Open
Abstract
Piwi proteins associate with piRNAs and functions in epigenetic programming, post-transcriptional regulation, transposon silencing, and germline development. However, it is not known whether the diverse functions of these proteins are molecularly separable. Here we report that Piwi interacts with Tudor-SN (Tudor staphylococcal nuclease, TSN) antagonistically in regulating spermatogenesis but synergistically in silencing transposons. However, it is not required for piRNA biogenesis. TSN is known to participate in diverse molecular functions such as RNAi, degradation of hyper-edited miRNAs, and spliceosome assembly. We show that TSN colocalizes with Piwi in primordial germ cells (PGCs) and embryonic somatic cells. In adult ovaries and testes, TSN is ubiquitously expressed and enriched in the cytoplasm of both germline and somatic cells. The tsn mutants display a higher mitotic index of spermatogonia, accumulation of spermatocytes, defects in meiotic cytokinesis, a decreased number of spermatids, and eventually reduced male fertility. Germline-specific TSN-expression analysis demonstrates that this function is germline-dependent. Different from other known Piwi interters, TSN represses Piwi expression at both protein and mRNA levels. Furthermore, reducing piwi expression in the germline rescues tsn mutant phenotype in a dosage-dependent manner, demonstrating that Piwi and TSN interact antagonistically in germ cells to regulate spermatogenesis. However, the tsn deficiency has little, if any, impact on piRNA biogenesis but displays a synergistic effect with piwi mutants in transposon de-silencing. Our results reveal the biological function of TSN and its contrasting modes of interaction with Piwi in spermatogenesis, transposon silencing, and piRNA biogenesis. Piwi proteins bind to a large class of small noncoding RNAs called Piwi-interacting RNAs (piRNAs). These proteins have emerged as major players in germline development, stem cell self-renewal, transposon silencing, and gene regulation. However, it is not known whether these functions of Piwi proteins represent separate molecular mechanisms. Furthermore, although multiple Piwi interactors have been identified, including Tudor-domain-containing proteins, none of them regulates Piwi expression or interacts with Piwi antagonistically, or only impact on a subset of Piwi functions. Here we show that Drosophila Piwi interacts with a special Tudor-domain-containing protein called Tudor-SN (Tudor staphylococcal nuclease, TSN). TSN is drastically different from the known Piwi interactors because it represses Piwi mRNA and protein expression and interacts with Piwi antagonistically in spermatogenesis but synergistically in transposon silencing. However, this interaction is not required for piRNA biogenesis. Our study represents the first demonstration that different functions of Piwi are mediated by different molecular mechanisms. In addition, this is the first in vivo study that reveals the biological function of TSN protein in an organism.
Collapse
Affiliation(s)
- Hsueh-Yen Ku
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Vamsi K. Gangaraju
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Hongying Qi
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Na Liu
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Haifan Lin
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
111
|
Abstract
Primordial germ cells are usually made early in the development of an organism. These are the mother of all stem cells that are necessary for propagation of the species, yet use highly diverse mechanisms between organisms. How they are specified, and when and where they form, are central to developmental biology. Using diverse organisms to study this development is illuminating for understanding the mechanics these cells use in this essential function and for identifying the breadth of evolutionary changes that have occurred between species. This essay emphasizes how echinoderms may contribute to the patchwork quilt of our understanding of germ line formation during embryogenesis.
Collapse
Affiliation(s)
- Gary M Wessel
- Department of Molecular and Cellular Biology, Brown University, Providence, Rhode Island, USA.
| |
Collapse
|
112
|
McFarlane RJ, Feichtinger J, Larcombe L. Cancer germline gene activation: friend or foe? Cell Cycle 2015; 13:2151-2. [PMID: 25111983 PMCID: PMC4111666 DOI: 10.4161/cc.29661] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
| | - Julia Feichtinger
- Institute for Knowledge Discovery; Graz University of Technology; Graz, Austria; Core Facility Bioinformatics; Austrian Centre of Industrial Biotechnology; Graz, Austria
| | - Lee Larcombe
- MRC Functional Genomics Unit; Department of Physiology, Anatomy and Genetics; University of Oxford; Oxford; UK
| |
Collapse
|
113
|
Yant L, Bomblies K. Genome management and mismanagement--cell-level opportunities and challenges of whole-genome duplication. Genes Dev 2015; 29:2405-19. [PMID: 26637526 PMCID: PMC4691946 DOI: 10.1101/gad.271072.115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Whole-genome duplication (WGD) doubles the DNA content in the nucleus and leads to polyploidy. In whole-organism polyploids, WGD has been implicated in adaptability and the evolution of increased genome complexity, but polyploidy can also arise in somatic cells of otherwise diploid plants and animals, where it plays important roles in development and likely environmental responses. As with whole organisms, WGD can also promote adaptability and diversity in proliferating cell lineages, although whether WGD is beneficial is clearly context-dependent. WGD is also sometimes associated with aging and disease and may be a facilitator of dangerous genetic and karyotypic diversity in tumorigenesis. Scaling changes can affect cell physiology, but problems associated with WGD in large part seem to arise from problems with chromosome segregation in polyploid cells. Here we discuss both the adaptive potential and problems associated with WGD, focusing primarily on cellular effects. We see value in recognizing polyploidy as a key player in generating diversity in development and cell lineage evolution, with intriguing parallels across kingdoms.
Collapse
Affiliation(s)
- Levi Yant
- John Innes Centre, Colney, Norwich NR4 7UH, United Kingdom
| | | |
Collapse
|
114
|
Lim RSM, Kai T. A piece of the pi(e): The diverse roles of animal piRNAs and their PIWI partners. Semin Cell Dev Biol 2015; 47-48:17-31. [PMID: 26582251 DOI: 10.1016/j.semcdb.2015.10.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Small non-coding RNAs are indispensable to many biological processes. A class of endogenous small RNAs, termed PIWI-interacting RNAs (piRNAs) because of their association with PIWI proteins, has known roles in safeguarding the genome against inordinate transposon mobilization, embryonic development, and stem cell regulation, among others. This review discusses the biogenesis of animal piRNAs and their diverse functions together with their PIWI protein partners, both in the germline and in somatic cells, and highlights the evolutionarily conserved aspects of these molecular players in animal biology.
Collapse
Affiliation(s)
- Robyn S M Lim
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.
| | - Toshie Kai
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.
| |
Collapse
|
115
|
Shen J, Lu J, Sui L, Wang D, Yin M, Hoffmann I, Legler A, Pflugfelder GO. The orthologous Tbx transcription factors Omb and TBX2 induce epithelial cell migration and extrusion in vivo without involvement of matrix metalloproteinases. Oncotarget 2015; 5:11998-2015. [PMID: 25344916 PMCID: PMC4322970 DOI: 10.18632/oncotarget.2426] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/02/2014] [Indexed: 01/06/2023] Open
Abstract
The transcription factors TBX2 and TBX3 are overexpressed in various human cancers. Here, we investigated the effect of overexpressing the orthologous Tbx genes Drosophila optomotor-blind (omb) and human TBX2 in the epithelium of the Drosophila wing imaginal disc and observed two types of cell motility. Omb/TBX2 overexpressing cells could move within the plane of the epithelium. Invasive cells migrated long-distance as single cells retaining or regaining normal cell shape and apico-basal polarity in spite of attenuated apical DE-cadherin concentration. Inappropriate levels of DE-cadherin were sufficient to drive cell migration in the wing disc epithelium. Omb/TBX2 overexpression and reduced DE-cadherin-dependent adhesion caused the formation of actin-rich lateral cell protrusions. Omb/TBX2 overexpressing cells could also delaminate basally, penetratingthe basal lamina, however, without degradation of extracellular matrix. Expression of Timp, an inhibitor of matrix metalloproteases, blocked neither intraepithelial motility nor basal extrusion. Our results reveal an MMP-independent mechanism of cell invasion and suggest a conserved role of Tbx2-related proteins in cell invasion and metastasis-related processes.
Collapse
Affiliation(s)
- Jie Shen
- Department of Entomology, China Agricultural University, Beijing, China
| | - Juan Lu
- Department of Entomology, China Agricultural University, Beijing, China
| | - Liyuan Sui
- Department of Entomology, China Agricultural University, Beijing, China
| | - Dan Wang
- Department of Entomology, China Agricultural University, Beijing, China
| | - Meizhen Yin
- Key Laboratory of Carbon Fiber and Functional Polymers, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Inka Hoffmann
- Institute of Genetics, Johannes Gutenberg-University, Mainz, Germany
| | - Anne Legler
- Institute of Genetics, Johannes Gutenberg-University, Mainz, Germany
| | | |
Collapse
|
116
|
Abstract
This protocol describes a method to allograft Drosophila larval tissue into adult fly hosts that can be used to assay the tumorigenic potential of mutant tissues. The tissue of interest is dissected, loaded into a fine glass needle and implanted into a host. Upon implantation, nontransformed tissues do not overgrow beyond their normal size, but malignant tumors grow without limit, are invasive and kill the host. By using this method, Drosophila malignant tumors can be transplanted repeatedly, for years, and therefore they can be aged beyond the short life span of flies. Because several hosts can be implanted using different pieces from a single tumor, the method also allows the tumor mass to be increased to facilitate further studies that may require large amounts of tissue (i.e., genomics, proteomics and so on). This method also provides an operational definition of hyperplastic, benign and malignant growth. The injection procedure itself requires only ∼1 d. Tumor development can then be monitored until the death of the implanted hosts.
Collapse
|
117
|
Abstract
Germ cells are the special cells in the body that undergo meiosis to generate gametes and subsequently entire new organisms after fertilization, a process that continues generation after generation. Recent studies have expanded our understanding of the factors and mechanisms that specify germ cell fate, including the partitioning of maternally supplied 'germ plasm', inheritance of epigenetic memory and expression of transcription factors crucial for primordial germ cell (PGC) development. Even after PGCs are specified, germline fate is labile and thus requires protective mechanisms, such as global transcriptional repression, chromatin state alteration and translation of only germline-appropriate transcripts. Findings from diverse species continue to provide insights into the shared and divergent needs of these special reproductive cells.
Collapse
Affiliation(s)
- Susan Strome
- Molecular, Cell &Developmental Biology, University of California Santa Cruz, Santa Cruz, California 95064, USA
| | - Dustin Updike
- Kathryn W. Davis Center for Regenerative Biology &Medicine, Mount Desert Island Biological Laboratory, Bar Harbor, Maine 04672, USA
| |
Collapse
|
118
|
EAG2 potassium channel with evolutionarily conserved function as a brain tumor target. Nat Neurosci 2015; 18:1236-46. [PMID: 26258683 DOI: 10.1038/nn.4088] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/15/2015] [Indexed: 12/15/2022]
Abstract
Over 20% of the drugs for treating human diseases target ion channels, but no cancer drug approved by the US Food and Drug Administration (FDA) is intended to target an ion channel. We found that the EAG2 (Ether-a-go-go 2) potassium channel has an evolutionarily conserved function for promoting brain tumor growth and metastasis, delineate downstream pathways, and uncover a mechanism for different potassium channels to functionally cooperate and regulate mitotic cell volume and tumor progression. EAG2 potassium channel was enriched at the trailing edge of migrating medulloblastoma (MB) cells to regulate local cell volume dynamics, thereby facilitating cell motility. We identified the FDA-approved antipsychotic drug thioridazine as an EAG2 channel blocker that reduces xenografted MB growth and metastasis, and present a case report of repurposing thioridazine for treating a human patient. Our findings illustrate the potential of targeting ion channels in cancer treatment.
Collapse
|
119
|
Zhang C, Robinson BS, Xu W, Yang L, Yao B, Zhao H, Byun PK, Jin P, Veraksa A, Moberg KH. The ecdysone receptor coactivator Taiman links Yorkie to transcriptional control of germline stem cell factors in somatic tissue. Dev Cell 2015; 34:168-80. [PMID: 26143992 DOI: 10.1016/j.devcel.2015.05.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 04/25/2015] [Accepted: 05/14/2015] [Indexed: 12/16/2022]
Abstract
The Hippo pathway is a conserved signaling cascade that modulates tissue growth. Although its core elements are well defined, factors modulating Hippo transcriptional outputs remain elusive. Here we show that components of the steroid-responsive ecdysone (Ec) pathway modulate Hippo transcriptional effects in imaginal disc cells. The Ec receptor coactivator Taiman (Tai) interacts with the Hippo transcriptional coactivator Yorkie (Yki) and promotes expression of canonical Yki-responsive genes. Tai enhances Yki-driven growth, while Tai loss, or a form of Tai unable to bind Yki, suppresses Yki-driven tissue growth. This growth suppression is not correlated with impaired induction of canonical Hippo-responsive genes but with suppression of a distinct pro-growth program of Yki-induced/Tai-dependent genes, including the germline stem cell factors nanos and piwi. These data reveal Hippo/Ec pathway crosstalk in the form a Yki-Tai complex that collaboratively induces germline genes as part of a transcriptional program that is normally repressed in developing somatic epithelia.
Collapse
Affiliation(s)
- Can Zhang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brian S Robinson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Wenjian Xu
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Liu Yang
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Heya Zhao
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Phil K Byun
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; Graduate Program in Genetics and Molecular Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Alexey Veraksa
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Kenneth H Moberg
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
120
|
Le Bescont A, Vitte AL, Debernardi A, Curtet S, Buchou T, Vayr J, de Reyniès A, Ito A, Guardiola P, Brambilla C, Yoshida M, Brambilla E, Rousseaux S, Khochbin S. Receptor-Independent Ectopic Activity of Prolactin Predicts Aggressive Lung Tumors and Indicates HDACi-Based Therapeutic Strategies. Antioxid Redox Signal 2015; 23:1-14. [PMID: 24512221 PMCID: PMC4492736 DOI: 10.1089/ars.2013.5581] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS Ectopic activation of tissue-specific genes accompanies malignant transformation in many cancers. Prolactin (PRL) aberrant activation in lung cancer was investigated here to highlight its value as a biomarker. RESULTS PRL is ectopically activated in a subset of very aggressive lung tumors, associated with a rapid fatal outcome, in our cohort of 293 lung tumor patients and in an external independent series of patients. Surprisingly PRL receptor expression was not detected in the vast majority of PRL-expressing lung tumors. Additionally, the analysis of the PRL transcripts in lung tumors and cell lines revealed systematic truncations of their 5' regions, including the signal peptide-encoding portions. PRL expression was found to sustain cancer-specific gene expression circuits encompassing genes that are normally responsive to hypoxia. Interestingly, this analysis also indicated that histone deacetylase (HDAC) inhibitors could counteract the PRL-associated transcriptional activity. INNOVATION AND CONCLUSION Altogether, this work not only unravels a yet unknown oncogenic mechanism but also indicates that the specific category of PRL-expressing aggressive lung cancers could be particularly responsive to an HDAC inhibitor-based treatment.
Collapse
Affiliation(s)
- Aurore Le Bescont
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | - Anne-Laure Vitte
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | - Alexandra Debernardi
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | - Sandrine Curtet
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | - Thierry Buchou
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | - Jessica Vayr
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | - Aurélien de Reyniès
- 2 Cartes d'Identité des Tumeurs Program, Ligue Nationale Contre le Cancer , Paris, France
| | - Akihiro Ito
- 3 Chemical Genetics Laboratory , RIKEN, Wako, Japan
| | - Philippe Guardiola
- 4 INSERM U892, Centre de Recherche sur le Cancer Nantes Angers and UMR_S 892, Université d'Angers , Plateforme SNP, Transcriptome & Epigénomique, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Christian Brambilla
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | | | - Elisabeth Brambilla
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | - Sophie Rousseaux
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| | - Saadi Khochbin
- 1 INSERM U823, Université Joseph Fourier-Grenoble 1 , Institut Albert Bonniot, Grenoble, France
| |
Collapse
|
121
|
Affiliation(s)
| | | | - Alastair S H Goldman
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, UK
| |
Collapse
|
122
|
A Forward Genetic Screen for Suppressors of Somatic P Granules in Caenorhabditis elegans. G3-GENES GENOMES GENETICS 2015; 5:2209-15. [PMID: 26100681 PMCID: PMC4593002 DOI: 10.1534/g3.115.019257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In Caenorhabditis elegans, germline expression programs are actively repressed in somatic tissue by components of the synMuv (synthetic multi-vulva) B chromatin remodeling complex, which include homologs of tumor suppressors Retinoblastoma (Rb/LIN-35) and Malignant Brain Tumor (MBT/LIN-61). However, the full scope of pathways that suppress germline expression in the soma is unknown. To address this, we performed a mutagenesis and screened for somatic expression of GFP-tagged PGL-1, a core P-granule nucleating protein. Eight alleles were isolated from 4000 haploid genomes. Five of these alleles exhibit a synMuv phenotype, whereas the remaining three were identified as hypomorphic alleles of known synMuv B genes, lin-13 and dpl-1. These findings suggest that most suppressors of germline programs in the soma of C. elegans are either required for viability or function through synMuv B chromatin regulation.
Collapse
|
123
|
Yajima M, Wessel GM. Essential elements for translation: the germline factor Vasa functions broadly in somatic cells. Development 2015; 142:1960-70. [PMID: 25977366 PMCID: PMC4460737 DOI: 10.1242/dev.118448] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 03/30/2015] [Indexed: 01/23/2023]
Abstract
Vasa is a conserved RNA-helicase found in the germ lines of all metazoans tested. Whereas Vasa presence is often indicated as a metric for germline determination in animals, it is also expressed in stem cells of diverse origin. Recent research suggests, however, that Vasa has a much broader function, including a significant role in cell cycle regulation. Results herein indicate that Vasa is utilized widely, and often induced transiently, during development in diverse somatic cells and adult precursor tissues. We identified that Vasa in the sea urchin is essential for: (1) general mRNA translation during embryogenesis, (2) developmental re-programming upon manipulations to the embryo and (3) larval wound healing. We also learned that Vasa interacted with mRNAs in the perinuclear area and at the spindle in an Importin-dependent manner during cell cycle progression. These results suggest that, when present, Vasa functions are essential to contributing to developmental regulation.
Collapse
Affiliation(s)
- Mamiko Yajima
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA
| | - Gary M Wessel
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA
| |
Collapse
|
124
|
Grelet S, Andries V, Polette M, Gilles C, Staes K, Martin AP, Kileztky C, Terryn C, Dalstein V, Cheng CW, Shen CY, Birembaut P, Van Roy F, Nawrocki-Raby B. The human NANOS3 gene contributes to lung tumour invasion by inducing epithelial-mesenchymal transition. J Pathol 2015; 237:25-37. [PMID: 25904364 DOI: 10.1002/path.4549] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/30/2015] [Accepted: 04/17/2015] [Indexed: 01/01/2023]
Abstract
We have explored the role of the human NANOS3 gene in lung tumour progression. We show that NANOS3 is over-expressed by invasive lung cancer cells and is a prognostic marker for non-small cell lung carcinomas (NSCLCs). NANOS3 gene expression is restricted in testis and brain and is regulated by epigenetic events. It is up-regulated in cultured cells undergoing epithelial - mesenchymal transition (EMT). NANOS3 over-expression in human NSCLC cell lines enhances their invasiveness by up-regulating EMT, whereas its silencing induces mesenchymal - epithelial transition. NANOS3 represses E-cadherin at the transcriptional level and up-regulates vimentin post-transcriptionally. Also, we show that NANOS3 binds mRNAs encoding vimentin and regulates the length of their poly(A) tail. Finally, NANOS3 can also protect vimentin mRNA from microRNA-mediated repression. We thus demonstrate a role for NANOS3 in the acquisition of invasiveness by human lung tumour cells and propose a new mechanism of post-transcriptional regulation of EMT.
Collapse
Affiliation(s)
- Simon Grelet
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France
| | - Vanessa Andries
- Department of Biomedical Molecular Biology, Ghent University, Belgium.,Molecular Cell Biology Unit, Inflammation Research Centre, Ghent, Belgium
| | - Myriam Polette
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France.,Laboratory of Histology, CHU Reims, France
| | - Christine Gilles
- Laboratory of Developmental and Tumour Biology, GIGA-Cancer, University of Liège, Belgium
| | - Katrien Staes
- Department of Biomedical Molecular Biology, Ghent University, Belgium.,Molecular Cell Biology Unit, Inflammation Research Centre, Ghent, Belgium
| | | | - Claire Kileztky
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France
| | - Christine Terryn
- Plateforme Imagerie Cellulaire et Tissulaire, University of Reims Champagne-Ardenne, France
| | - Véronique Dalstein
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France.,Laboratory of Histology, CHU Reims, France
| | - Chun-Wen Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Philippe Birembaut
- INSERM UMR-S 903, SFR CAP-Santé, University of Reims Champagne-Ardenne, France.,Laboratory of Histology, CHU Reims, France
| | - Frans Van Roy
- Department of Biomedical Molecular Biology, Ghent University, Belgium.,Molecular Cell Biology Unit, Inflammation Research Centre, Ghent, Belgium
| | | |
Collapse
|
125
|
Tan Y, Liu L, Liao M, Zhang C, Hu S, Zou M, Gu M, Li X. Emerging roles for PIWI proteins in cancer. Acta Biochim Biophys Sin (Shanghai) 2015; 47:315-324. [PMID: 25854579 DOI: 10.1093/abbs/gmv018] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/13/2015] [Indexed: 12/18/2022] Open
Abstract
It is generally accepted that PIWI proteins are predominately expressed in the germline but absent in somatic tissues. Their best-characterized role is to suppress transposon expression, which ensures genomic stability in the germline. However, increasing evidence has suggested that PIWI proteins are linked to the hallmarks of cancer defined by Weinberg and Hanahan, such as cell proliferation, anti-apoptosis, genomic instability, invasion and metastasis. This provides new possibilities for anticancer therapies through the targeting of PIWI proteins, which may have fewer side effects due to their potential classification as a CTA (cancer/testis antigen). Furthermore, PIWI has been proposed to act as a diagnostic and prognostic marker for many types of cancer, and even to differentiate early- and late-stage cancers. We herein summarize the latest progress in this exciting field, hoping to encourage new investigations of PIWIs in cancer biology that will help to develop new therapeutics for clinical application.
Collapse
Affiliation(s)
- Yi Tan
- Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou 325035, China
| | - Lianyong Liu
- Department of Endocrine, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai 200135, China
| | - Mingan Liao
- College of Horticulture, Sichuan Agricultural University, Ya'an 625014, China
| | - Chaobao Zhang
- Department of Endocrine, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai 200135, China
| | - Shuanggang Hu
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Mei Zou
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Mingjun Gu
- Department of Endocrine, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai 200135, China
| | - Xiangqi Li
- Department of Endocrine, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai 200135, China
| |
Collapse
|
126
|
piRNA involvement in genome stability and human cancer. J Hematol Oncol 2015; 8:38. [PMID: 25895683 PMCID: PMC4412036 DOI: 10.1186/s13045-015-0133-5] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/31/2015] [Indexed: 12/16/2022] Open
Abstract
PIWI-interacting RNAs (piRNAs) are a large family of small, single-stranded, non-coding RNAs present throughout the animal kingdom. They form complexes with several members of the PIWI clade of Argonaute proteins and carry out regulatory functions. Their best established biological role is the inhibition of transposon mobilization, which they enforce both at the transcriptional level, through regulation of heterochromatin formation, and by promoting transcript degradation. In this capacity, piRNAs and PIWI proteins are at the heart of the germline cells’ efforts to preserve genome integrity. Additional regulatory roles of piRNAs and PIWI proteins in gene expression are becoming increasingly apparent. PIWI proteins and piRNAs are often detected in human cancers deriving from germline cells as well as somatic tissues. Their detection in cancer correlates with poorer clinical outcomes, suggesting that they play a functional role in the biology of cancer. Nonetheless, the currently available information, while highly suggestive, is still not sufficient to entirely discriminate between a ‘passenger’ role for the ectopic expression of piRNAs and PIWI proteins in cancer from a ‘driver’ role in the pathogenesis of these diseases. In this article, we review some of the key available evidence for the role of piRNAs and PIWI in human cancer and discuss ways in which our understanding of their functions may be improved.
Collapse
|
127
|
Peschansky VJ, Wahlestedt C. Non-coding RNAs as direct and indirect modulators of epigenetic regulation. Epigenetics 2015; 9:3-12. [PMID: 24739571 DOI: 10.4161/epi.27473] [Citation(s) in RCA: 366] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epigenetic regulation of gene expression is an increasingly well-understood concept that explains much of the contribution of an organism's environment and experience to its biology. However, discussion persists as to which mechanisms can be classified as epigenetic. Ongoing research continues to uncover novel pathways, including the important role of non-protein coding RNA transcripts in epigenetic gene regulation. We know that the majority of human and other mammalian transcripts are not translated but that many of these are nonetheless functional. These non-coding RNAs (ncRNAs) can be short (<200 nt) or long (<200 nt) and are further classified by genomic origin and mechanism of action. We discuss examples of ncRNAs that interact with histone modifying complexes or DNA methyltransferases to regulate gene expression, others that are targets of these epigenetic mechanisms, and propose a model in which such transcripts feed back into an epigenetic regulatory network.
Collapse
Affiliation(s)
- Veronica J Peschansky
- Center for Therapeutic Innovation & Department of Psychiatry and Behavioral Sciences; University of Miami; Miller School of Medicine; Miami, FL USA
| | - Claes Wahlestedt
- Center for Therapeutic Innovation & Department of Psychiatry and Behavioral Sciences; University of Miami; Miller School of Medicine; Miami, FL USA
| |
Collapse
|
128
|
Hasegawa K, Sin HS, Maezawa S, Broering TJ, Kartashov AV, Alavattam KG, Ichijima Y, Zhang F, Bacon WC, Greis KD, Andreassen PR, Barski A, Namekawa SH. SCML2 establishes the male germline epigenome through regulation of histone H2A ubiquitination. Dev Cell 2015; 32:574-88. [PMID: 25703348 PMCID: PMC4391279 DOI: 10.1016/j.devcel.2015.01.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 12/23/2014] [Accepted: 01/16/2015] [Indexed: 01/03/2023]
Abstract
Gametogenesis is dependent on the expression of germline-specific genes. However, it remains unknown how the germline epigenome is distinctly established from that of somatic lineages. Here we show that genes commonly expressed in somatic lineages and spermatogenesis-progenitor cells undergo repression in a genome-wide manner in late stages of the male germline and identify underlying mechanisms. SCML2, a germline-specific subunit of a Polycomb repressive complex 1 (PRC1), establishes the unique epigenome of the male germline through two distinct antithetical mechanisms. SCML2 works with PRC1 and promotes RNF2-dependent ubiquitination of H2A, thereby marking somatic/progenitor genes on autosomes for repression. Paradoxically, SCML2 also prevents RNF2-dependent ubiquitination of H2A on sex chromosomes during meiosis, thereby enabling unique epigenetic programming of sex chromosomes for male reproduction. Our results reveal divergent mechanisms involving a shared regulator by which the male germline epigenome is distinguished from that of the soma and progenitor cells.
Collapse
Affiliation(s)
- Kazuteru Hasegawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Ho-Su Sin
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - So Maezawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Tyler J Broering
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Andrey V Kartashov
- Division of Allergy and Immunology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Kris G Alavattam
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Yosuke Ichijima
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Fan Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - W Clark Bacon
- Division of Allergy and Immunology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Kenneth D Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Paul R Andreassen
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Artem Barski
- Division of Allergy and Immunology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Satoshi H Namekawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA.
| |
Collapse
|
129
|
Abstract
PIWI-interacting RNAs (piRNAs) are a class of small RNAs that are 24-31 nucleotides in length. They associate with PIWI proteins, which constitute a germline-specific subclade of the Argonaute family, to form effector complexes known as piRNA-induced silencing complexes, which repress transposons via transcriptional or posttranscriptional mechanisms and maintain germline genome integrity. In addition to having a role in transposon silencing, piRNAs in diverse organisms function in the regulation of cellular genes. In some cases, piRNAs have shown transgenerational inheritance to pass on the memory of "self" and "nonself," suggesting a contribution to various cellular processes over generations. Many piRNA factors have been identified; however, both the molecular mechanisms leading to the production of mature piRNAs and the effector phases of gene silencing are still enigmatic. Here, we summarize the current state of our knowledge on the biogenesis of piRNA, its biological functions, and the underlying mechanisms.
Collapse
Affiliation(s)
- Yuka W Iwasaki
- Department of Molecular Biology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | | | | |
Collapse
|
130
|
González C, Sedwick C. Cayetano González: mothers, daughters, stemness, and cancer. ACTA ACUST UNITED AC 2015; 208:254-5. [PMID: 25646082 PMCID: PMC4315249 DOI: 10.1083/jcb.2083pi] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
González studies centrosomal inheritance, asymmetric cell division, and cancer.
Collapse
|
131
|
Davie K, Jacobs J, Atkins M, Potier D, Christiaens V, Halder G, Aerts S. Discovery of transcription factors and regulatory regions driving in vivo tumor development by ATAC-seq and FAIRE-seq open chromatin profiling. PLoS Genet 2015; 11:e1004994. [PMID: 25679813 PMCID: PMC4334524 DOI: 10.1371/journal.pgen.1004994] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 01/08/2015] [Indexed: 01/24/2023] Open
Abstract
Genomic enhancers regulate spatio-temporal gene expression by recruiting specific combinations of transcription factors (TFs). When TFs are bound to active regulatory regions, they displace canonical nucleosomes, making these regions biochemically detectable as nucleosome-depleted regions or accessible/open chromatin. Here we ask whether open chromatin profiling can be used to identify the entire repertoire of active promoters and enhancers underlying tissue-specific gene expression during normal development and oncogenesis in vivo. To this end, we first compare two different approaches to detect open chromatin in vivo using the Drosophila eye primordium as a model system: FAIRE-seq, based on physical separation of open versus closed chromatin; and ATAC-seq, based on preferential integration of a transposon into open chromatin. We find that both methods reproducibly capture the tissue-specific chromatin activity of regulatory regions, including promoters, enhancers, and insulators. Using both techniques, we screened for regulatory regions that become ectopically active during Ras-dependent oncogenesis, and identified 3778 regions that become (over-)activated during tumor development. Next, we applied motif discovery to search for candidate transcription factors that could bind these regions and identified AP-1 and Stat92E as key regulators. We validated the importance of Stat92E in the development of the tumors by introducing a loss of function Stat92E mutant, which was sufficient to rescue the tumor phenotype. Additionally we tested if the predicted Stat92E responsive regulatory regions are genuine, using ectopic induction of JAK/STAT signaling in developing eye discs, and observed that similar chromatin changes indeed occurred. Finally, we determine that these are functionally significant regulatory changes, as nearby target genes are up- or down-regulated. In conclusion, we show that FAIRE-seq and ATAC-seq based open chromatin profiling, combined with motif discovery, is a straightforward approach to identify functional genomic regulatory regions, master regulators, and gene regulatory networks controlling complex in vivo processes.
Collapse
Affiliation(s)
- Kristofer Davie
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Jelle Jacobs
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Mardelle Atkins
- Laboratory of Growth Control and Cancer Research, Center for Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for the Biology of Disease, Laboratory for Molecular Cancer Biology, Leuven, Belgium
| | - Delphine Potier
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Valerie Christiaens
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Georg Halder
- Laboratory of Growth Control and Cancer Research, Center for Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for the Biology of Disease, Laboratory for Molecular Cancer Biology, Leuven, Belgium
| | - Stein Aerts
- Laboratory of Computational Biology, Center for Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
132
|
Mukherjee S, Kong J, Brat DJ. Cancer stem cell division: when the rules of asymmetry are broken. Stem Cells Dev 2014; 24:405-16. [PMID: 25382732 DOI: 10.1089/scd.2014.0442] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Asymmetric division of stem cells is a highly conserved and tightly regulated process by which a single stem cell produces two daughter cells and simultaneously directs the differential fate of both: one retains its stem cell identity while the other becomes specialized and loses stem cell properties. Coordinating these events requires control over numerous intra- and extracellular biological processes and signaling networks. In the initial stages, critical events include the compartmentalization of fate determining proteins within the mother cell and their subsequent passage to the appropriate daughter cell. Disturbance of these events results in an altered dynamic of self-renewing and differentiation within the cell population, which is highly relevant to the growth and progression of cancer. Other critical events include proper asymmetric spindle assembly, extrinsic regulation through micro-environmental cues, and noncanonical signaling networks that impact cell division and fate determination. In this review, we discuss mechanisms that maintain the delicate balance of asymmetric cell division in normal tissues and describe the current understanding how some of these mechanisms are deregulated in cancer. The universe is asymmetric and I am persuaded that life, as it is known to us, is a direct result of the asymmetry of the universe or of its indirect consequences. The universe is asymmetric. -Louis Pasteur.
Collapse
Affiliation(s)
- Subhas Mukherjee
- 1 Department of Pathology and Laboratory Medicine, Emory University , Atlanta, Georgia
| | | | | |
Collapse
|
133
|
Homem CCF, Steinmann V, Burkard TR, Jais A, Esterbauer H, Knoblich JA. Ecdysone and mediator change energy metabolism to terminate proliferation in Drosophila neural stem cells. Cell 2014; 158:874-888. [PMID: 25126791 DOI: 10.1016/j.cell.2014.06.024] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/10/2014] [Accepted: 06/06/2014] [Indexed: 11/19/2022]
Abstract
Stem cells are highly abundant during early development but become a rare population in most adult organs. The molecular mechanisms causing stem cells to exit proliferation at a specific time are not well understood. Here, we show that changes in energy metabolism induced by the steroid hormone ecdysone and the Mediator initiate an irreversible cascade of events leading to cell-cycle exit in Drosophila neural stem cells. We show that the timely induction of oxidative phosphorylation and the mitochondrial respiratory chain are required in neuroblasts to uncouple the cell cycle from cell growth. This results in a progressive reduction in neuroblast cell size and ultimately in terminal differentiation. Brain tumor mutant neuroblasts fail to undergo this shrinkage process and continue to proliferate until adulthood. Our findings show that cell size control can be modified by systemic hormonal signaling and reveal a unique connection between metabolism and proliferation in stem cells.
Collapse
Affiliation(s)
- Catarina C F Homem
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030 Vienna, Austria
| | - Victoria Steinmann
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030 Vienna, Austria
| | - Thomas R Burkard
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030 Vienna, Austria
| | - Alexander Jais
- Department of Laboratory Medicine, Medical University Vienna, 1090 Vienna, Austria
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University Vienna, 1090 Vienna, Austria
| | - Juergen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030 Vienna, Austria.
| |
Collapse
|
134
|
Identification of a class of human cancer germline genes with transcriptional silencing refractory to the hypomethylating drug 5-aza-2'-deoxycytidine. Oncoscience 2014; 1:745-50. [PMID: 25594001 PMCID: PMC4278271 DOI: 10.18632/oncoscience.95] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 11/10/2014] [Indexed: 12/20/2022] Open
Abstract
Bona fide germline genes have expression restricted to the germ cells of the gonads. Testis-specific germline development-associated genes can become activated in cancer cells and can potentially drive the oncogenic process and serve as therapeutic/biomarker targets; such germline genes are referred to as cancer/testis genes. Many cancer/testis genes are silenced via hypermethylation of CpG islands in their associated transcriptional control regions and become activated upon treatment with DNA hypomethylating agents; such hypomethylation-induced activation of cancer/testis genes provides a potential combination approach to augment immunotherapeutics. Thus, understanding cancer/testis gene regulation is of increasing clinical importance. Previously studied cancer/testis gene activation has focused on X chromosome encoded cancer/testis genes. Here we find that a sub-set of non-X encoded cancer/testis genes are silenced in non-germline cells via a mechanism that is refractory to epigenetic dysregulation, including treatment with the hypomethylating agent 5-aza-2'-deoxycytidine and the histone deacetylase inhibitor tricostatin A. These findings formally indicate that there is a sub-group of the clinically important cancer/testis genes that are unlikely to be activated in clinical therapeutic approaches using hypomethylating agents and it indicates a unique transcriptional silencing mechanism for germline genes in non-germline cells that might provide a target mechanism for new clinical therapies.
Collapse
|
135
|
Meier K, Brehm A. Chromatin regulation: how complex does it get? Epigenetics 2014; 9:1485-95. [PMID: 25482055 PMCID: PMC4622878 DOI: 10.4161/15592294.2014.971580] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/18/2014] [Accepted: 09/29/2014] [Indexed: 12/16/2022] Open
Abstract
Gene transcription is tightly regulated at different levels to ensure that the transcriptome of the cell is appropriate for developmental stage and cell type. The chromatin state in which a gene is embedded determines its expression level to a large extent. Activation or repression of transcription is typically accomplished by the recruitment of chromatin-associated multisubunit protein complexes that combine several molecular tools, such as histone-binding and chromatin-modifying activities. Recent biochemical purifications of such complexes have revealed a substantial diversity. On the one hand, complexes that were thought to be unique have been revealed to be part of large complex families. On the other hand, protein subunits that were thought to only exist in separate complexes have been shown to coexist in novel assemblies. In this review we discuss our current knowledge of repressor complexes that contain MBT domain proteins and/or the CoREST co-repressor and use them as a paradigm to illustrate the unexpected heterogeneity and tool sharing of chromatin regulating protein complexes. These recent insights also challenge the ways we define and think about protein complexes in general.
Collapse
Key Words
- ATP, adenosine triphosphate
- BAP, brahma associated protein
- BHC80, BRAF-histone deacetylase complex 80
- BRG1, brahma Related Gene 1
- CHD, chromo domain helicase DNA binding
- CoREST
- CoREST REST, corepressor
- DNA, deoxyribonucleic acid
- DNMT, DNA methyltransferase
- DP-1, dimerization partner 1
- E2F, E2 transcription Factor
- ELM2, EGL-27 and MTA1 homology 2
- ES cell, embryonic stem cells
- H, histone
- HDAC, histone deacetylas
- HMTase, histone methylase
- HP1, heterochromatin protein 1
- K, lysine
- L3MBTL, lethal 3 malignant brain tumor-like
- LINT, l(3)mbt interacting
- LSD1, lysine-specific demethylase 1
- Lint-1, l(3)mbt interacting 1
- MBT protein
- MBT, malignant brain tumor
- MBTS, malignant brain tumor signature
- NPA1, nucleosome assembly protein
- NRSF, neural-restrictive silencing factor
- NuRD, nucleosome remodeling and deacetylase
- PBAP, polybromo-associated BAP
- PHD, plant homeo domain
- PRC1, polycomb repressive complex 1
- PRE, polycomb responsive element
- Pc, polycomb
- PcG, polycomb group
- Ph, polyhomeotic
- Pho, pleiohomeotic
- PhoRC, Pho repressive complex
- Psc, posterior sex combs
- RB, retinoblastoma
- REST, repressor element 1 silencing transcription factor
- RNA, ribonucleic acid
- Rpd3, reduced potassium dependency 3
- SANT, SWI/ADA2/N-CoR/TFIIIB
- SCML, sex combs on midleg-like
- SLC, SFMBT1, LSD1, CoREST
- SWH, Salvador-Warts-Hippo
- SWI/SNF, switching defective/sucrose non-fermenting
- Sce, sex combs extra
- Scm, sex combs on midleg
- Sfmbt, Scm-related gene containing 4 mbt domains
- TSS, transcription start site
- YY1, ying-yang 1
- ZNF, zinc finger
- complex family
- dL(3)mbt, Drosophila Lethal 3 malignant brain tumor
- hBRM, human Brahma
- l(3)mbt, lethal 3 malignant brain tumor
- protein complex
- transcriptional regulation
Collapse
Affiliation(s)
- Karin Meier
- Institut für Molekularbiologie und Tumorforschung; Philipps-Universität Marburg; Marburg, Germany
- Instituto de Fisiología Celular; Departamento de Genética Molecular; Universidad Nacional Autónoma de México; México City, México
| | - Alexander Brehm
- Institut für Molekularbiologie und Tumorforschung; Philipps-Universität Marburg; Marburg, Germany
| |
Collapse
|
136
|
Wang J, Li J, Santana-Santos L, Shuda M, Sobol RW, Van Houten B, Qian W. A novel strategy for targeted killing of tumor cells: Induction of multipolar acentrosomal mitotic spindles with a quinazolinone derivative mdivi-1. Mol Oncol 2014; 9:488-502. [PMID: 25458053 DOI: 10.1016/j.molonc.2014.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 10/07/2014] [Accepted: 10/08/2014] [Indexed: 01/05/2023] Open
Abstract
Traditional antimitotic drugs for cancer chemotherapy often have undesired toxicities to healthy tissues, limiting their clinical application. Developing novel agents that specifically target tumor cell mitosis is needed to minimize the toxicity and improve the efficacy of this class of anticancer drugs. We discovered that mdivi-1 (mitochondrial division inhibitor-1), which was originally reported as an inhibitor of mitochondrial fission protein Drp1, specifically disrupts M phase cell cycle progression only in human tumor cells, but not in non-transformed fibroblasts or epithelial cells. The antimitotic effect of mdivi-1 is Drp1 independent, as mdivi-1 induces M phase abnormalities in both Drp1 wild-type and Drp1 knockout SV40-immortalized/transformed MEF cells. We also identified that the tumor transformation process required for the antimitotic effect of mdivi-1 is downstream of SV40 large T and small t antigens, but not hTERT-mediated immortalization. Mdivi-1 induces multipolar mitotic spindles in tumor cells regardless of their centrosome numbers. Acentrosomal spindle poles, which do not contain the bona-fide centrosome components γ-tubulin and centrin-2, were found to contribute to the spindle multipolarity induced by mdivi-1. Gene expression profiling revealed that the genes involved in oocyte meiosis and assembly of acentrosomal microtubules are highly expressed in tumor cells. We further identified that tumor cells have enhanced activity in the nucleation and assembly of acentrosomal kinetochore-attaching microtubules. Mdivi-1 inhibited the integration of acentrosomal microtubule-organizing centers into centrosomal asters, resulting in the development of acentrosomal mitotic spindles preferentially in tumor cells. The formation of multipolar acentrosomal spindles leads to gross genome instability and Bax/Bak-dependent apoptosis. Taken together, our studies indicate that inducing multipolar spindles composing of acentrosomal poles in mitosis could achieve tumor-specific antimitotic effect, and mdivi-1 thus represents a novel class of compounds as acentrosomal spindle inducers (ASI).
Collapse
Affiliation(s)
- Jingnan Wang
- Tsinghua University School of Medicine, Tsinghua University, Haidian District, Beijing 100084, China
| | - Jianfeng Li
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine and Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | - Lucas Santana-Santos
- Biomedical Informatics, and Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Masahiro Shuda
- Molecular Virology Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | - Robert W Sobol
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine and Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15213, USA
| | - Bennett Van Houten
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine and Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | - Wei Qian
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine and Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| |
Collapse
|
137
|
Chromatin reader L(3)mbt requires the Myb-MuvB/DREAM transcriptional regulatory complex for chromosomal recruitment. Proc Natl Acad Sci U S A 2014; 111:E4234-43. [PMID: 25249635 DOI: 10.1073/pnas.1416321111] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Lethal malignant brain tumors (lmbt) result from the loss of the conserved transcriptional repressor l(3)mbt, in Drosophila melanogaster. Similar mutations in the human homolog L3MBTL1 correlate with some cancers. The protein's C-terminal MBT repeats bind mono and dimethylated histones in vitro, which could influence recruitment of L3MBTL1 to its target sites. The L(3)mbt chromatin targeting mechanism, however, is controversial and several studies suggest insufficiency or a minor role for histone methylation in determining the site specificity for recruitment. We report that L(3)mbt colocalizes with core members of the Myb-MuvB/DREAM (MMB/DREAM) transcriptional regulatory complex genome-wide, and that L(3)mbt-mediated repression requires this complex in salivary glands and larval brains. Loss of l(3)mbt or of MMB components through mutation cause similar spurious expression of genes, including the transposon regulatory gene piwi, in terminally differentiated cells. The DNA-binding MMB core component Mip120 (Lin54) is required for L(3)mbt recruitment to chromosomes, whereas Mip130 (Lin9) (an MMB core protein) and E2f2 (an MMB transcriptional repressor) are not, but are essential for repression. Cytolocalization experiments suggest the presence of site-specific differential composition of MMB in polytene chromosomes where some loci were bound by a Myb-containing or alternatively, an E2f2 and L(3)mbt form of the complex.
Collapse
|
138
|
Dnmt3b Prefers Germ Line Genes and Centromeric Regions: Lessons from the ICF Syndrome and Cancer and Implications for Diseases. BIOLOGY 2014; 3:578-605. [PMID: 25198254 PMCID: PMC4192629 DOI: 10.3390/biology3030578] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 07/18/2014] [Accepted: 08/21/2014] [Indexed: 01/06/2023]
Abstract
The correct establishment and maintenance of DNA methylation patterns are critical for mammalian development and the control of normal cell growth and differentiation. DNA methylation has profound effects on the mammalian genome, including transcriptional repression, modulation of chromatin structure, X chromosome inactivation, genomic imprinting, and the suppression of the detrimental effects of repetitive and parasitic DNA sequences on genome integrity. Consistent with its essential role in normal cells and predominance at repetitive genomic regions, aberrant changes of DNA methylation patterns are a common feature of diseases with chromosomal and genomic instabilities. In this context, the functions of DNA methyltransferases (DNMTs) can be affected by mutations or alterations of their expression. DNMT3B, which is involved in de novo methylation, is of particular interest not only because of its important role in development, but also because of its dysfunction in human diseases. Expression of catalytically inactive isoforms has been associated with cancer risk and germ line hypomorphic mutations with the ICF syndrome (Immunodeficiency Centromeric instability Facial anomalies). In these diseases, global genomic hypomethylation affects repeated sequences around centromeric regions, which make up large blocks of heterochromatin, and is associated with chromosome instability, impaired chromosome segregation and perturbed nuclear architecture. The review will focus on recent data about the function of DNMT3B, and the consequences of its deregulated activity on pathological DNA hypomethylation, including the illicit activation of germ line-specific genes and accumulation of transcripts originating from repeated satellite sequences, which may represent novel physiopathological biomarkers for human diseases. Notably, we focus on cancer and the ICF syndrome, pathological contexts in which hypomethylation has been extensively characterized. We also discuss the potential contribution of these deregulated protein-coding and non-coding transcription programs to the perturbation of cellular phenotypes.
Collapse
|
139
|
Huang T, Alvarez A, Hu B, Cheng SY. Noncoding RNAs in cancer and cancer stem cells. CHINESE JOURNAL OF CANCER 2014; 32:582-93. [PMID: 24206916 PMCID: PMC3845549 DOI: 10.5732/cjc.013.10170] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In recent years, it has become increasingly apparent that noncoding RNAs (ncRNA) are of crucial importance for human cancer. The functional relevance of ncRNAs is particularly evident for microRNAs (miRNAs) and long noncoding RNAs (lncRNAs). miRNAs are endogenously expressed small RNA sequences that act as post-transcriptional regulators of gene expression and have been extensively studied for their roles in cancers, whereas lncRNAs are emerging as important players in the cancer paradigm in recent years. These noncoding genes are often aberrantly expressed in a variety of human cancers. However, the biological functions of most ncRNAs remain largely unknown. Recently, evidence has begun to accumulate describing how ncRNAs are dysregulated in cancer and cancer stem cells, a subset of cancer cells harboring self-renewal and differentiation capacities. These studies provide insight into the functional roles that ncRNAs play in tumor initiation, progression, and resistance to therapies, and they suggest ncRNAs as attractive therapeutic targets and potentially useful diagnostic tools.
Collapse
Affiliation(s)
- Tianzhi Huang
- The Ken & Ruth Davee Department of Neuro-logy, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA. ,
| | | | | | | |
Collapse
|
140
|
Drosophila neuroblasts as a new model for the study of stem cell self-renewal and tumour formation. Biosci Rep 2014; 34:BSR20140008. [PMID: 24965943 PMCID: PMC4114065 DOI: 10.1042/bsr20140008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Drosophila larval brain stem cells (neuroblasts) have emerged as an important model for the study of stem cell asymmetric division and the mechanisms underlying the transformation of neural stem cells into tumour-forming cancer stem cells. Each Drosophila neuroblast divides asymmetrically to produce a larger daughter cell that retains neuroblast identity, and a smaller daughter cell that is committed to undergo differentiation. Neuroblast self-renewal and differentiation are tightly controlled by a set of intrinsic factors that regulate ACD (asymmetric cell division). Any disruption of these two processes may deleteriously affect the delicate balance between neuroblast self-renewal and progenitor cell fate specification and differentiation, causing neuroblast overgrowth and ultimately lead to tumour formation in the fly. In this review, we discuss the mechanisms underlying Drosophila neural stem cell self-renewal and differentiation. Furthermore, we highlight emerging evidence in support of the notion that defects in ACD in mammalian systems, which may play significant roles in the series of pathogenic events leading to the development of brain cancers.
Collapse
|
141
|
Abstract
PURPOSE OF REVIEW Systematic genetic and epigenetic alterations occurring in almost all cancer cells result in the ectopic expression of a variety of tissue-specific potent regulatory factors. This review sheds light on a new aspect of cancer based on the integration of the 'out of context' activity of tissue-restricted genes into the biology of cancer cells. RECENT FINDINGS A systematic screen for the ectopic activation of tissue-restricted genes in a variety of cancers has revealed that many normally silent genes are expressed in tumours of all origins. This aberrant gene activation not only could be used as a source of biomarkers, but also, in several cases, reveals clear oncogenic mechanisms associated with the corresponding ectopically expressed factors. SUMMARY The characteristic of all cancer cells, which systematically reprogram tissue-specific gene expression and activate silent genes, can be exploited to develop new anticancer strategies aiming at the detection of malignant states, the prediction of their evolution and drug sensitivity and the discovery of new therapeutic approaches.
Collapse
|
142
|
Vinnitsky V. The development of a malignant tumor is due to a desperate asexual self-cloning process in which cancer stem cells develop the ability to mimic the genetic program of germline cells. INTRINSICALLY DISORDERED PROTEINS 2014; 2:e29997. [PMID: 28232878 PMCID: PMC5314931 DOI: 10.4161/idp.29997] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 02/08/2023]
Abstract
To date there is no explanation why the development of almost all types of solid tumors occurs sharing a similar scenario: (1) creation of a cancer stem cell (CSC), (2) CSC multiplication and formation of a multicellular tumor spheroid (TS), (3) vascularization of the TS and its transformation into a vascularized primary tumor, (4) metastatic spreading of CSCs, (5) formation of a metastatic TSs and its transformation into metastatic tumors, and (6) potentially endless repetition of this cycle of events. The above gaps in our knowledge are related to the biology of cancer and specifically to tumorigenesis, which covers the process from the creation of a CSC to the formation of a malignant tumor and the development of metastases. My Oncogerminative Theory of Tumorigenesis considers tumor formation as a dynamic self-organizing process that mimics a self-organizing process of early embryo development. In the initial step in that process, gene mutations combined with epigenetic dysregulation cause somatic cells to be reprogrammed into CSCs, which are immortal pseudo-germline cells. Mimicking the behavior of fertilized germline cells, the CSC achieves immortality by passing through the stages of its life-cycle and developing into a pseudo-blastula-stage embryo, which manifests in the body as a malignant tumor. In this view, the development of a malignant tumor from a CSC is a phenomenon of developmental biology, which we named a desperate asexual self-cloning event. The theory explains seven core characteristics of malignant tumors: (1) CSC immortality, (2) multistep development of a malignant tumor from a single CSC, (3) heterogeneity of malignant tumor cell populations, (4) metastatic spread of CSCs, (5) invasive growth, (6) malignant progression, and (7) selective immune tolerance toward cancer cells. The Oncogerminative Theory of Tumorigenesis suggests new avenues for discovery of revolutionary therapies to treat, prevent, and eradicate cancer.
Collapse
Affiliation(s)
- Vladimir Vinnitsky
- Department of Experimental Cancer Therapeutics; R.E. Kavetsky Institute for Experimental Pathology, Oncology, and Radiobiology; Kiev, Ukraine
- Sequent Development (CRO), LLC; Madison, VA USA
| |
Collapse
|
143
|
Liu C, Ma Z, Xu S, Hou J, Hu Y, Yu Y, Liu R, Chen Z, Lu Y. Activation of the germ-cell potential of human bone marrow-derived cells by a chemical carcinogen. Sci Rep 2014; 4:5564. [PMID: 24998261 PMCID: PMC4083294 DOI: 10.1038/srep05564] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 06/17/2014] [Indexed: 01/06/2023] Open
Abstract
Embryonic/germ cell traits are common in malignant tumors and are thought to be involved in malignant tumor behaviors. The reasons why tumors show strong embryonic/germline traits (displaced germ cells or gametogenic programming reactivation) are controversial. Here, we show that a chemical carcinogen, 3-methyl-cholanthrene (3-MCA), can trigger the germ-cell potential of human bone marrow-derived cells (hBMDCs). 3-MCA promoted the generation of germ cell-like cells from induced hBMDCs that had undergone malignant transformation, whereas similar results were not observed in the parallel hBMDC culture at the same time point. The malignant transformed hBMDCs spontaneously and more efficiently generated into germ cell-like cells even at the single-cell level. The germ cell-like cells from induced hBMDCs were similar to natural germ cells in many aspects, including morphology, gene expression, proliferation, migration, further development, and teratocarcinoma formation. Therefore, our results demonstrate that a chemical carcinogen can reactivate the germline phenotypes of human somatic tissue-derived cells, which might provide a novel idea to tumor biology and therapy.
Collapse
Affiliation(s)
- Chunfang Liu
- 1] Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China [2]
| | - Zhan Ma
- 1] Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China [2] Department of Laboratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China [3]
| | - Songtao Xu
- 1] Department of Thoracic Surgery, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, China [2]
| | - Jun Hou
- Department of Pathology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yao Hu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Yinglu Yu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Ruilai Liu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zhihong Chen
- Department of Pulmonary Medicine, Research Institute of Respiratory Disease, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuan Lu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| |
Collapse
|
144
|
Glycerol-3-phosphate acyltranferase-2 behaves as a cancer testis gene and promotes growth and tumorigenicity of the breast cancer MDA-MB-231 cell line. PLoS One 2014; 9:e100896. [PMID: 24967918 PMCID: PMC4072688 DOI: 10.1371/journal.pone.0100896] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 06/01/2014] [Indexed: 02/06/2023] Open
Abstract
The de novo synthesis of glycerolipids in mammalian cells begins with the acylation of glycerol-3-phosphate, catalyzed by glycerol-3-phosphate acyltransferase (GPAT). GPAT2 is a mitochondrial isoform primarily expressed in testis under physiological conditions. Because it is aberrantly expressed in multiple myeloma, it has been proposed as a novel cancer testis gene. Using a bioinformatics approach, we found that GPAT2 is highly expressed in melanoma, lung, prostate and breast cancer, and we validated GPAT2 expression at the protein level in breast cancer by immunohistochemistry. In this case GPAT2 expression correlated with a higher histological grade. 5-Aza-2′ deoxycytidine treatment of human cells lines induced GPAT2 expression suggesting epigenetic regulation of gene expression. In order to evaluate the contribution of GPAT2 to the tumor phenotype, we silenced its expression in MDA-MB-231 cells. GPAT2 knockdown diminished cell proliferation, anchorage independent growth, migration and tumorigenicity, and increased staurosporine-induced apoptosis. In contrast, GPAT2 over-expression increased cell proliferation rate and resistance to staurosporine-induced apoptosis. To understand the functional role of GPAT2, we performed a co-expression analysis in mouse and human testis and found a significant association with semantic terms involved in cell cycle, DNA integrity maintenance, piRNA biogenesis and epigenetic regulation. Overall, these results indicate the GPAT2 would be directly associated with the control of cell proliferation. In conclusion, we confirm GPAT2 as a cancer testis gene and that its expression contributes to the tumor phenotype of MDA-MB-231 cells.
Collapse
|
145
|
Ku HY, Lin H. PIWI proteins and their interactors in piRNA biogenesis, germline development and gene expression. Natl Sci Rev 2014; 1:205-218. [PMID: 25512877 DOI: 10.1093/nsr/nwu014] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PIWI-interacting RNAs (piRNAs) are a complex class of small non-coding RNAs that are mostly 24-32 nucleotides in length and composed of at least hundreds of thousands of species that specifically interact with the PIWI protein subfamily of the ARGONAUTE family. Recent studies revealed that PIWI proteins interact with a number of proteins, especially the TUDOR-domain-containing proteins, to regulate piRNA biogenesis and regulatory function. Current research also provides evidence that PIWI proteins and piRNAs are not only crucial for transposon silencing in the germline, but also mediate novel mechanisms of epigenetic programming, DNA rearrangements, mRNA turnover, and translational control both in the germline and in the soma. These new discoveries begin to reveal an exciting new dimension of gene regulation in the cell.
Collapse
Affiliation(s)
- Hsueh-Yen Ku
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Haifan Lin
- Yale Stem Cell Center and Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
146
|
van Wolfswinkel JC. Piwi and Potency: PIWI Proteins in Animal Stem Cells and Regeneration. Integr Comp Biol 2014; 54:700-13. [DOI: 10.1093/icb/icu084] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
147
|
Lim SL, Ricciardelli C, Oehler MK, De Arao Tan IMD, Russell D, Grützner F. Overexpression of piRNA pathway genes in epithelial ovarian cancer. PLoS One 2014; 9:e99687. [PMID: 24932571 PMCID: PMC4059699 DOI: 10.1371/journal.pone.0099687] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 05/19/2014] [Indexed: 11/18/2022] Open
Abstract
The importance of the Piwi-interacting RNA (piRNA) pathway for germ cell maintenance, genome integrity, DNA methylation and retrotransposon control raises possible roles of this pathway in cancer. Indeed aberrant expression of human PIWI orthologs and Maelstrom has been observed in various cancers. In this study we explored the expression and function of piRNA pathway genes in human ovarian cancer, based on our recent work, which showed widespread expression of piRNA pathway genes in the mammalian. Our work shows that PIWIL1 and MAEL expression is significantly increased in malignant EOC (n = 25) compared to benign tumor tissues (n = 19) and normal ovarian tissue (n = 8). The expression of PIWIL3 is lower in malignant and benign tissues when compared to normal ovary. Sequencing of PIWIL1 transcript revealed that in many tumors deletion of exon 17 leads to the introduction of a premature stop codon in the PIWI domain, likely due to a splicing error. In situ hybridization on tumor sections revealed that L1, PIWIL1, 2 and MAEL are specifically expressed in epithelial cells (cancerous cells) of EOC. Furthermore, PIWIL2 and MAEL are co-expressed in the stromal cells adjacent to tumor cells. Since PIWIL1 and MAEL are up regulated in malignant EOC and expressed in the epithelial cells, we investigated if these two genes affect invasiveness of ovarian cancer cell lines that do not normally express these genes. PIWIL1 and MAEL were transiently over expressed in the ovarian cancer cell line SKOV3, followed by real-time measurements of cell invasiveness. Surprisingly both PIWIL1 and MAEL over expression decreased the invasiveness of SKOV3 cells. Our findings support a growing body of evidence that shows that genes in this pathway are upregulated in cancer. In ovarian cancer we show for the first time that Piwil1 transcript may often be abnormal result in non functional product. In contrast to what has been observed in other cell types, we found that PIWIL1 and MAEL have a repressive effect on cell invasiveness.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Argonaute Proteins/genetics
- Argonaute Proteins/metabolism
- Base Sequence
- Carcinoma, Ovarian Epithelial
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line, Tumor
- DNA-Binding Proteins
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- In Situ Hybridization
- Middle Aged
- Molecular Sequence Data
- Neoplasm Invasiveness
- Neoplasm Staging
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/pathology
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- Ovary/metabolism
- Ovary/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction/genetics
- Transcription Factors
- Transfection
Collapse
Affiliation(s)
- Shu Ly Lim
- Robinson Research Institute, School of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Martin K. Oehler
- Discipline of Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Izza M. D. De Arao Tan
- Discipline of Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Darryl Russell
- Discipline of Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Frank Grützner
- Robinson Research Institute, School of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
- * E-mail:
| |
Collapse
|
148
|
Functional analysis of the Drosophila embryonic germ cell transcriptome by RNA interference. PLoS One 2014; 9:e98579. [PMID: 24896584 PMCID: PMC4045815 DOI: 10.1371/journal.pone.0098579] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 05/05/2014] [Indexed: 11/19/2022] Open
Abstract
In Drosophila melanogaster, primordial germ cells are specified at the posterior pole of the very early embryo. This process is regulated by the posterior localized germ plasm that contains a large number of RNAs of maternal origin. Transcription in the primordial germ cells is actively down-regulated until germ cell fate is established. Bulk expression of the zygotic genes commences concomitantly with the degradation of the maternal transcripts. Thus, during embryogenesis, maternally provided and zygotically transcribed mRNAs determine germ cell development collectively. In an effort to identify novel genes involved in the regulation of germ cell behavior, we carried out a large-scale RNAi screen targeting both maternal and zygotic components of the embryonic germ line transcriptome. We identified 48 genes necessary for distinct stages in germ cell development. We found pebble and fascetto to be essential for germ cell migration and germ cell division, respectively. Our data uncover a previously unanticipated role of mei-P26 in maintenance of embryonic germ cell fate. We also performed systematic co-RNAi experiments, through which we found a low rate of functional redundancy among homologous gene pairs. As our data indicate a high degree of evolutionary conservation in genetic regulation of germ cell development, they are likely to provide valuable insights into the biology of the germ line in general.
Collapse
|
149
|
Huang Y, Bai JY, Ren HT. piRNA biogenesis and its functions. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2014. [DOI: 10.1134/s1068162014030169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
150
|
Feichtinger J, Larcombe L, McFarlane RJ. Meta-analysis of expression of l(3)mbt tumor-associated germline genes supports the model that a soma-to-germline transition is a hallmark of human cancers. Int J Cancer 2014; 134:2359-65. [PMID: 24243547 PMCID: PMC4166677 DOI: 10.1002/ijc.28577] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/27/2013] [Accepted: 10/18/2013] [Indexed: 12/14/2022]
Abstract
Evidence is starting to emerge indicating that tumorigenesis in metazoans involves a soma-to-germline transition, which may contribute to the acquisition of neoplastic characteristics. Here, we have meta-analyzed gene expression profiles of the human orthologs of Drosophila melanogaster germline genes that are ectopically expressed in l(3)mbt brain tumors using gene expression datasets derived from a large cohort of human tumors. We find these germline genes, some of which drive oncogenesis in D. melanogaster, are similarly ectopically activated in a wide range of human cancers. Some of these genes normally have expression restricted to the germline, making them of particular clinical interest. Importantly, these analyses provide additional support to the emerging model that proposes a soma-to-germline transition is a general hallmark of a wide range of human tumors. This has implications for our understanding of human oncogenesis and the development of new therapeutic and biomarker targets with clinical potential.
Collapse
Affiliation(s)
- Julia Feichtinger
- North West Cancer Research Institute, Bangor University, Brambell Building, Bangor, Gwynedd, United Kingdom; Institute for Genomics and Bioinformatics, Graz University of Technology, Graz, Austria
| | | | | |
Collapse
|