1
|
Huang YT, Calvi BR. Activation of a Src-JNK pathway in unscheduled endocycling cells of the Drosophila wing disc induces a chronic wounding response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642788. [PMID: 40161657 PMCID: PMC11952448 DOI: 10.1101/2025.03.12.642788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The endocycle is a specialized cell cycle during which cells undergo repeated G / S phases to replicate DNA without division, leading to large polyploid cells. The transition from a mitotic cycle to an endocycle can be triggered by various stresses, which results in unscheduled, or induced endocycling cells (iECs). While iECs can be beneficial for wound healing, they can also be detrimental by impairing tissue growth or promoting cancer. However, the regulation of endocycling and its role in tissue growth remain poorly understood. Using the Drosophila wing disc as a model, we previously demonstrated that iEC growth is arrested through a Jun N-Terminal Kinase (JNK)-dependent, reversible senescence-like response. However, it remains unclear how JNK is activated in iECs and how iECs impact overall tissue structure. In this study, we performed a genetic screen and identified the Src42A-Shark-Slpr pathway as an upstream regulator of JNK in iECs, leading to their senescence-like arrest. We found that tissues recognize iECs as wounds, releasing wound-related signals that induce a JNK-dependent developmental delay. Similar to wound closure, this response triggers Src-JNK-mediated actomyosin remodeling, yet iECs persist rather than being eliminated. Our findings suggest that the tissue response to iECs shares key signaling and cytoskeletal regulatory mechanisms with wound healing and dorsal closure, a developmental process during Drosophila embryogenesis. However, because iECs are retained within the tissue, they create a unique system that may serve as a model for studying chronic wounds and tumor progression.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Indiana University, Bloomington, Indiana, 47405 USA
| | - Brian R. Calvi
- Department of Biology, Indiana University, Bloomington, Indiana, 47405 USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, 46202 USA
| |
Collapse
|
2
|
Standring S, Heckenhauer J, Stewart RJ, Frandsen PB. Unraveling the genetics of underwater caddisfly silk. Trends Genet 2025:S0168-9525(25)00004-6. [PMID: 39893090 DOI: 10.1016/j.tig.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 02/04/2025]
Abstract
Hundreds of thousands of arthropod species use silk to capture prey, build protective structures, or anchor eggs. While most silk-producers are terrestrial, caddisflies construct silken capture nets and portable cases in aquatic environments. Given the potential practical applications of this underwater bioadhesive, there is an emerging body of research focused on understanding the evolution of the genetic architecture of aquatic silk. This research has unveiled molecular adaptations specific to caddisfly silk, such as extensive phosphorylation of the primary silk protein and the existence of numerous unique accessory silk proteins. We discuss the molecular evolution of caddisfly silk genes, how they interact with the environment, and suggest future directions for caddisfly silk genetics research.
Collapse
Affiliation(s)
- Samantha Standring
- Department of Plant and Wildlife Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Jacqueline Heckenhauer
- Senckenberg Research Institute and Natural History Museum Frankfurt, Terrestrial Zoology, 60325 Frankfurt am Main, Germany; LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), 60325 Frankfurt am Main, Germany
| | - Russell J Stewart
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Paul B Frandsen
- Department of Plant and Wildlife Sciences, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
3
|
von Saucken VE, Windner SE, Armetta G, Baylies MK. Postsynaptic BMP signaling regulates myonuclear properties in Drosophila larval muscles. J Cell Biol 2025; 224:e202404052. [PMID: 39475469 PMCID: PMC11530350 DOI: 10.1083/jcb.202404052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/13/2024] [Accepted: 10/13/2024] [Indexed: 11/04/2024] Open
Abstract
The syncytial mammalian muscle fiber contains a heterogeneous population of (myo)nuclei. At the neuromuscular junction (NMJ), myonuclei have specialized positioning and gene expression. However, it remains unclear how myonuclei are recruited and what regulates myonuclear output at the NMJ. Here, we identify specific properties of myonuclei located near the Drosophila larval NMJ. These synaptic myonuclei have increased size in relation to their surrounding cytoplasmic domain (size scaling), increased DNA content (ploidy), and increased levels of transcription factor pMad, a readout for BMP signaling activity. Our genetic manipulations show that local BMP signaling affects muscle size, nuclear size, ploidy, and NMJ size and function. In support, RNA sequencing analysis reveals that pMad regulates genes involved in muscle growth, ploidy (i.e., E2f1), and neurotransmission. Our data suggest that muscle BMP signaling instructs synaptic myonuclear output that positively shapes the NMJ synapse. This study deepens our understanding of how myonuclear heterogeneity supports local signaling demands to fine tune cellular function and NMJ activity.
Collapse
Affiliation(s)
- Victoria E. von Saucken
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell-Rockefeller-Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
- Biochemistry, Cell and Developmental Biology, and Molecular Biology (BCMB) Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Stefanie E. Windner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giovanna Armetta
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary K. Baylies
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
4
|
Yang Q, Wijaya F, Kapoor R, Chandrasekaran H, Jagtiani S, Moran I, Hime GR. Unusual modes of cell and nuclear divisions characterise Drosophila development. Biochem Soc Trans 2024; 52:2281-2295. [PMID: 39508395 PMCID: PMC11668308 DOI: 10.1042/bst20231341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
The growth and development of metazoan organisms is dependent upon a co-ordinated programme of cellular proliferation and differentiation, from the initial formation of the zygote through to maintenance of mature organs in adult organisms. Early studies of proliferation of ex vivo cultures and unicellular eukaryotes described a cyclic nature of cell division characterised by periods of DNA synthesis (S-phase) and segregation of newly synthesized chromosomes (M-phase) interspersed by seeming inactivity, the gap phases, G1 and G2. We now know that G1 and G2 play critical roles in regulating the cell cycle, including monitoring of favourable environmental conditions to facilitate cell division, and ensuring genomic integrity prior to DNA replication and nuclear division. M-phase is usually followed by the physical separation of nascent daughters, termed cytokinesis. These phases where G1 leads to S phase, followed by G2 prior to M phase and the subsequent cytokinesis to produce two daughters, both identical in genomic composition and cellular morphology are what might be termed an archetypal cell division. Studies of development of many different organs in different species have demonstrated that this stereotypical cell cycle is often subverted to produce specific developmental outcomes, and examples from over 100 years of analysis of the development of Drosophila melanogaster have uncovered many different modes of cell division within this one species.
Collapse
Affiliation(s)
- Qiaolin Yang
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Fernando Wijaya
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Ridam Kapoor
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Harshaa Chandrasekaran
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Siddhant Jagtiani
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Izaac Moran
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gary R. Hime
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
5
|
Huang YT, Hesting LL, Calvi BR. An unscheduled switch to endocycles induces a reversible senescent arrest that impairs growth of the Drosophila wing disc. PLoS Genet 2024; 20:e1011387. [PMID: 39226333 PMCID: PMC11398662 DOI: 10.1371/journal.pgen.1011387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/13/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogaster wing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| | - Lauren L Hesting
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| | - Brian R Calvi
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
6
|
Butler K, Ahmed S, Jablonski J, Hookway TA. Engineered Cardiac Microtissue Biomanufacturing Using Human Induced Pluripotent Stem Cell Derived Epicardial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593960. [PMID: 38798424 PMCID: PMC11118268 DOI: 10.1101/2024.05.13.593960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Epicardial cells are a crucial component in constructing in vitro 3D tissue models of the human heart, contributing to the ECM environment and the resident mesenchymal cell population. Studying the human epicardium and its development from the proepicardial organ is difficult, but induced pluripotent stem cells can provide a source of human epicardial cells for developmental modeling and for biomanufacturing heterotypic cardiac tissues. This study shows that a robust population of epicardial cells (approx. 87.7% WT1+) can be obtained by small molecule modulation of the Wnt signaling pathway. The population maintains WT1 expression and characteristic epithelial morphology over successive passaging, but increases in size and decreases in cell number, suggesting a limit to their expandability in vitro. Further, low passage number epicardial cells formed into more robust 3D microtissues compared to their higher passage counterparts, suggesting that the ideal time frame for use of these epicardial cells for tissue engineering and modeling purposes is early on in their differentiated state. Additionally, the differentiated epicardial cells displayed two distinct morphologic sub populations with a subset of larger, more migratory cells which led expansion of the epicardial cells across various extracellular matrix environments. When incorporated into a mixed 3D co-culture with cardiomyocytes, epicardial cells promoted greater remodeling and migration without impairing cardiomyocyte function. This study provides an important characterization of stem cell-derived epicardial cells, identifying key characteristics that influence their ability to fabricate consistent engineered cardiac tissues.
Collapse
Affiliation(s)
- Kirk Butler
- Biomedical Engineering Department, Binghamton University, the State University of New York, Binghamton NY 13902
| | - Saif Ahmed
- Biomedical Engineering Department, Binghamton University, the State University of New York, Binghamton NY 13902
| | - Justin Jablonski
- Biomedical Engineering Department, University of Rochester, Rochester, NY14627
| | - Tracy A. Hookway
- Biomedical Engineering Department, Binghamton University, the State University of New York, Binghamton NY 13902
| |
Collapse
|
7
|
von Saucken VE, Windner SE, Baylies MK. Postsynaptic BMP signaling regulates myonuclear properties in Drosophila larval muscles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588944. [PMID: 38645063 PMCID: PMC11030338 DOI: 10.1101/2024.04.10.588944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The syncytial mammalian muscle fiber contains a heterogeneous population of (myo)nuclei. At the neuromuscular junction (NMJ), myonuclei have specialized positioning and gene expression. However, it remains unclear how myonuclei are recruited and what regulates myonuclear output at the NMJ. Here, we identify specific properties of myonuclei located near the Drosophila larval NMJ. These synaptic myonuclei have increased size in relation to their surrounding cytoplasmic domain (scaling), increased DNA content (ploidy), and increased levels of transcription factor pMad, a readout for BMP signaling activity. Our genetic manipulations show local BMP signaling affects muscle size, nuclear size, ploidy, and NMJ size and function. In support, RNA sequencing analysis reveals that pMad regulates genes involved in muscle growth, ploidy (i.e., E2f1), and neurotransmission. Our data suggest that muscle BMP signaling instructs synaptic myonuclear output that then positively shapes the NMJ synapse. This study deepens our understanding of how myonuclear heterogeneity supports local signaling demands to fine tune cellular function and NMJ activity.
Collapse
Affiliation(s)
- Victoria E. von Saucken
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
- Weill Cornell-Rockefeller-Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065 USA
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065 USA
| | - Stefanie E. Windner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Mary K. Baylies
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| |
Collapse
|
8
|
Lim SE, Vicente-Munuera P, Mao Y. Forced back into shape: Mechanics of epithelial wound repair. Curr Opin Cell Biol 2024; 87:102324. [PMID: 38290420 DOI: 10.1016/j.ceb.2024.102324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
Wound repair, the closing of a hole, is inherently a physical process that requires the change of shape of materials, in this case, cells and tissues. Not only is efficient and accurate wound repair critical for restoring barrier function and reducing infection, but it is also critical for restoring the complex three-dimensional architecture of an organ. This re-sculpting of tissues requires the complex coordination of cell behaviours in multiple dimensions, in space and time, to ensure that the repaired structure can continue functioning optimally. Recent evidence highlights the importance of cell and tissue mechanics in 2D and 3D to achieve such seamless wound repair.
Collapse
Affiliation(s)
- Shu En Lim
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK
| | - Pablo Vicente-Munuera
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK
| | - Yanlan Mao
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Institute for the Physics of Living Systems, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
9
|
Herriage HC, Huang YT, Calvi BR. The antagonistic relationship between apoptosis and polyploidy in development and cancer. Semin Cell Dev Biol 2024; 156:35-43. [PMID: 37331841 PMCID: PMC10724375 DOI: 10.1016/j.semcdb.2023.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/20/2023]
Abstract
One of the important functions of regulated cell death is to prevent cells from inappropriately acquiring extra copies of their genome, a state known as polyploidy. Apoptosis is the primary cell death mechanism that prevents polyploidy, and defects in this apoptotic response can result in polyploid cells whose subsequent error-prone chromosome segregation are a major contributor to genome instability and cancer progression. Conversely, some cells actively repress apoptosis to become polyploid as part of normal development or regeneration. Thus, although apoptosis prevents polyploidy, the polyploid state can actively repress apoptosis. In this review, we discuss progress in understanding the antagonistic relationship between apoptosis and polyploidy in development and cancer. Despite recent advances, a key conclusion is that much remains unknown about the mechanisms that link apoptosis to polyploid cell cycles. We suggest that drawing parallels between the regulation of apoptosis in development and cancer could help to fill this knowledge gap and lead to more effective therapies.
Collapse
Affiliation(s)
- Hunter C Herriage
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Yi-Ting Huang
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Brian R Calvi
- Department of Biology, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
10
|
Huang YT, Hesting LL, Calvi BR. An unscheduled switch to endocycles induces a reversible senescent arrest that impairs growth of the Drosophila wing disc. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.585098. [PMID: 38559130 PMCID: PMC10980049 DOI: 10.1101/2024.03.14.585098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogasterwing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| | - Lauren L. Hesting
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| | - Brian R. Calvi
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| |
Collapse
|
11
|
White J, Hutson MS, Page-McCaw A. Wounding increases nuclear ploidy in wound-proximal epidermal cells of the Drosophila pupal notum. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001067. [PMID: 38495588 PMCID: PMC10943363 DOI: 10.17912/micropub.biology.001067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
After injury, tissues must replace cell mass and genome copy number. The mitotic cycle is one mechanism for replacement, but non-mitotic strategies have been observed in quiescent tissues to restore tissue ploidy after wounding. Here we report that nuclei of the mitotically capable Drosophila pupal notum enlarged following nearby laser ablation. Measuring DNA content, we determined that nuclei within 100 µm of a laser-wound increased their ploidy to ~8C, consistent with one extra S-phase. These data indicate non-mitotic repair strategies are not exclusively utilized by quiescent tissues and may be an underexplored wound repair strategy in mitotic tissues.
Collapse
Affiliation(s)
- James White
- Dept. Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
- Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - M. Shane Hutson
- Dept. Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, United States
- Dept. Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States
| | - Andrea Page-McCaw
- Dept. Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
- Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
12
|
Wang Y, Tamori Y. Polyploid Cancer Cell Models in Drosophila. Genes (Basel) 2024; 15:96. [PMID: 38254985 PMCID: PMC10815460 DOI: 10.3390/genes15010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Cells with an abnormal number of chromosomes have been found in more than 90% of solid tumors, and among these, polyploidy accounts for about 40%. Polyploidized cells most often have duplicate centrosomes as well as genomes, and thus their mitosis tends to promote merotelic spindle attachments and chromosomal instability, which produces a variety of aneuploid daughter cells. Polyploid cells have been found highly resistant to various stress and anticancer therapies, such as radiation and mitogenic inhibitors. In other words, common cancer therapies kill proliferative diploid cells, which make up the majority of cancer tissues, while polyploid cells, which lurk in smaller numbers, may survive. The surviving polyploid cells, prompted by acute environmental changes, begin to mitose with chromosomal instability, leading to an explosion of genetic heterogeneity and a concomitant cell competition and adaptive evolution. The result is a recurrence of the cancer during which the tenacious cells that survived treatment express malignant traits. Although the presence of polyploid cells in cancer tissues has been observed for more than 150 years, the function and exact role of these cells in cancer progression has remained elusive. For this reason, there is currently no effective therapeutic treatment directed against polyploid cells. This is due in part to the lack of suitable experimental models, but recently several models have become available to study polyploid cells in vivo. We propose that the experimental models in Drosophila, for which genetic techniques are highly developed, could be very useful in deciphering mechanisms of polyploidy and its role in cancer progression.
Collapse
Affiliation(s)
| | - Yoichiro Tamori
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| |
Collapse
|
13
|
Yadav V, Mishra R, Das P, Arya R. Cut homeodomain transcription factor is a novel regulator of growth and morphogenesis of cortex glia niche around neural cells. Genetics 2024; 226:iyad173. [PMID: 37751321 PMCID: PMC11491519 DOI: 10.1093/genetics/iyad173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Cortex glia in Drosophila central nervous system form a niche around neural cells for necessary signals to establish cross talk with their surroundings. These cells grow and expand their thin processes around neural cell bodies. Although essential for the development and function of the nervous system, how these cells make extensive and intricate connected networks remains largely unknown. In this study, we show that Cut, a homeodomain transcription factor, directly regulates the fate of the cortex glia, impacting neural stem cell (NSC) homeostasis. Focusing on the thoracic ventral nerve cord, we found that Cut is required for the normal growth and development of cortex glia and timely increase in DNA content through endocycle to later divide via acytokinetic mitosis. Knockdown of Cut in cortex glia significantly reduces the growth of cellular processes, the network around NSCs, and their progeny's cell bodies. Conversely, overexpression of Cut induces overall growth of the main processes at the expense of side ones. Whereas the Cut knockdown slows down the timely increase of DNA, the Cut overexpression results in a significant increase in nuclear size and volume and a 3-fold increase in DNA content of cortex glia. Further, we note that constitutively high Cut also interfered with nuclei separation during acytokinetic mitosis. Since the cortex glia form syncytial networks around neural cells, the finding identifies Cut as a novel regulator of glial growth and variant cell cycles to support a functional nervous system.
Collapse
Affiliation(s)
- Vaishali Yadav
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Ramkrishna Mishra
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Papri Das
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Richa Arya
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
14
|
Dehn AS, Duhaime L, Gogna N, Nishina PM, Kelley K, Losick VP. Epithelial mechanics are maintained by inhibiting cell fusion with age in Drosophila. J Cell Sci 2023; 136:jcs260974. [PMID: 37732459 PMCID: PMC10651104 DOI: 10.1242/jcs.260974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023] Open
Abstract
A characteristic of normal aging and age-related diseases is the remodeling of the cellular organization of a tissue through polyploid cell growth. Polyploidy arises from an increase in nuclear ploidy or the number of nuclei per cell. However, it is not known whether age-induced polyploidy is an adaption to stressors or a precursor to degeneration. Here, we find that abdominal epithelium of the adult fruit fly becomes polyploid with age through generation of multinucleated cells by cell fusion. Inhibition of fusion does not improve the lifespan of the fly, but does enhance its biomechanical fitness, a measure of the healthspan of the animal. Remarkably, Drosophila can maintain their epithelial tension and abdominal movements with age when cell fusion is inhibited. Epithelial cell fusion also appears to be dependent on a mechanical cue, as knockdown of Rho kinase, E-cadherin or α-catenin is sufficient to induce multinucleation in young animals. Interestingly, mutations in α-catenin in mice result in retina pigment epithelial multinucleation associated with macular disease. Therefore, we have discovered that polyploid cells arise by cell fusion and contribute to the decline in the biomechanical fitness of the animal with age.
Collapse
Affiliation(s)
- Ari S. Dehn
- Boston College, 140 Commonwealth Ave, Chestnut Hill, MA 02467, USA
| | - Levi Duhaime
- Boston College, 140 Commonwealth Ave, Chestnut Hill, MA 02467, USA
| | - Navdeep Gogna
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Patsy M. Nishina
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Kristina Kelley
- Boston College, 140 Commonwealth Ave, Chestnut Hill, MA 02467, USA
| | - Vicki P. Losick
- Boston College, 140 Commonwealth Ave, Chestnut Hill, MA 02467, USA
| |
Collapse
|
15
|
Sanz-Gómez N, González-Álvarez M, De Las Rivas J, de Cárcer G. Whole-Genome Doubling as a source of cancer: how, when, where, and why? Front Cell Dev Biol 2023; 11:1209136. [PMID: 37342233 PMCID: PMC10277508 DOI: 10.3389/fcell.2023.1209136] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/24/2023] [Indexed: 06/22/2023] Open
Abstract
Chromosome instability is a well-known hallmark of cancer, leading to increased genetic plasticity of tumoral cells, which favors cancer aggressiveness, and poor prognosis. One of the main sources of chromosomal instability are events that lead to a Whole-Genome Duplication (WGD) and the subsequently generated cell polyploidy. In recent years, several studies showed that WGD occurs at the early stages of cell transformation, which allows cells to later become aneuploid, thus leading to cancer progression. On the other hand, other studies convey that polyploidy plays a tumor suppressor role, by inducing cell cycle arrest, cell senescence, apoptosis, and even prompting cell differentiation, depending on the tissue cell type. There is still a gap in understanding how cells that underwent WGD can overcome the deleterious effect on cell fitness and evolve to become tumoral. Some laboratories in the chromosomal instability field recently explored this paradox, finding biomarkers that modulate polyploid cells to become oncogenic. This review brings a historical view of how WGD and polyploidy impact cell fitness and cancer progression, and bring together the last studies that describe the genes helping cells to adapt to polyploidy.
Collapse
Affiliation(s)
- Natalia Sanz-Gómez
- Cell Cycle and Cancer Biomarkers Laboratory, Cancer Biology Department, Instituto de Investigaciones Biomédicas “Alberto Sols“. (IIBM) CSIC-UAM, Madrid, Spain
| | - María González-Álvarez
- Cell Cycle and Cancer Biomarkers Laboratory, Cancer Biology Department, Instituto de Investigaciones Biomédicas “Alberto Sols“. (IIBM) CSIC-UAM, Madrid, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IBMCC), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca (USAL), Salamanca, Spain
| | - Guillermo de Cárcer
- Cell Cycle and Cancer Biomarkers Laboratory, Cancer Biology Department, Instituto de Investigaciones Biomédicas “Alberto Sols“. (IIBM) CSIC-UAM, Madrid, Spain
| |
Collapse
|
16
|
Enomoto M, Igaki T. Cell-cell interactions that drive tumorigenesis in Drosophila. Fly (Austin) 2022; 16:367-381. [PMID: 36413374 PMCID: PMC9683056 DOI: 10.1080/19336934.2022.2148828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cell-cell interactions within tumour microenvironment play crucial roles in tumorigenesis. Genetic mosaic techniques available in Drosophila have provided a powerful platform to study the basic principles of tumour growth and progression via cell-cell communications. This led to the identification of oncogenic cell-cell interactions triggered by endocytic dysregulation, mitochondrial dysfunction, cell polarity defects, or Src activation in Drosophila imaginal epithelia. Such oncogenic cooperations can be caused by interactions among epithelial cells, mesenchymal cells, and immune cells. Moreover, microenvironmental factors such as nutrients, local tissue structures, and endogenous growth signalling activities critically affect tumorigenesis. Dissecting various types of oncogenic cell-cell interactions at the single-cell level in Drosophila will greatly increase our understanding of how tumours progress in living animals.
Collapse
Affiliation(s)
- Masato Enomoto
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho, Kyoto, Japan
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho, Kyoto, Japan,CONTACT Tatsushi Igaki
| |
Collapse
|
17
|
De Chiara L, Conte C, Semeraro R, Diaz-Bulnes P, Angelotti ML, Mazzinghi B, Molli A, Antonelli G, Landini S, Melica ME, Peired AJ, Maggi L, Donati M, La Regina G, Allinovi M, Ravaglia F, Guasti D, Bani D, Cirillo L, Becherucci F, Guzzi F, Magi A, Annunziato F, Lasagni L, Anders HJ, Lazzeri E, Romagnani P. Tubular cell polyploidy protects from lethal acute kidney injury but promotes consequent chronic kidney disease. Nat Commun 2022; 13:5805. [PMID: 36195583 PMCID: PMC9532438 DOI: 10.1038/s41467-022-33110-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 09/02/2022] [Indexed: 11/09/2022] Open
Abstract
Acute kidney injury (AKI) is frequent, often fatal and, for lack of specific therapies, can leave survivors with chronic kidney disease (CKD). We characterize the distribution of tubular cells (TC) undergoing polyploidy along AKI by DNA content analysis and single cell RNA-sequencing. Furthermore, we study the functional roles of polyploidization using transgenic models and drug interventions. We identify YAP1-driven TC polyploidization outside the site of injury as a rapid way to sustain residual kidney function early during AKI. This survival mechanism comes at the cost of senescence of polyploid TC promoting interstitial fibrosis and CKD in AKI survivors. However, targeting TC polyploidization after the early AKI phase can prevent AKI-CKD transition without influencing AKI lethality. Senolytic treatment prevents CKD by blocking repeated TC polyploidization cycles. These results revise the current pathophysiological concept of how the kidney responds to acute injury and identify a novel druggable target to improve prognosis in AKI survivors. Acute kidney injury is frequent, often fatal and can leave survivors with chronic kidney disease. Here the authors show that tubular cell polyploidy reduces early fatality sustaining residual function but promotes chronic kidney disease, which can be prevented by blocking YAP1
Collapse
Affiliation(s)
- Letizia De Chiara
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Carolina Conte
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50139, Italy
| | - Paula Diaz-Bulnes
- Translational immunology, Instituto de Investigación Sanitaria del Principado de Asturias ISPA, 33011, Oviedo, Asturias, España
| | - Maria Lucia Angelotti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Benedetta Mazzinghi
- Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, 50139, Italy
| | - Alice Molli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy.,Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, 50139, Italy
| | - Giulia Antonelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Samuela Landini
- Medical Genetics Unit, Meyer Children's University Hospital, Florence, 50139, Italy
| | - Maria Elena Melica
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Anna Julie Peired
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50139, Italy
| | - Marta Donati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Gilda La Regina
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Marco Allinovi
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, Florence, 50134, Italy
| | - Fiammetta Ravaglia
- Nephrology and Dialysis Unit, Santo Stefano Hospital, Prato, 59100, Italy
| | - Daniele Guasti
- Department of Experimental & Clinical Medicine, Imaging Platform, University of Florence, Florence, 50139, Italy
| | - Daniele Bani
- Department of Experimental & Clinical Medicine, Imaging Platform, University of Florence, Florence, 50139, Italy
| | - Luigi Cirillo
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy.,Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, 50139, Italy
| | - Francesca Becherucci
- Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, 50139, Italy
| | - Francesco Guzzi
- Nephrology and Dialysis Unit, Santo Stefano Hospital, Prato, 59100, Italy
| | - Alberto Magi
- Department of Information Engineering, University of Florence, Florence, 50139, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, 50139, Italy.,Flow Cytometry Diagnostic Center and Immunotherapy (CDCI), Careggi University Hospital, Florence, 50134, Italy
| | - Laura Lasagni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, LMU Hospital, Munich, 80336, Germany
| | - Elena Lazzeri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy.
| | - Paola Romagnani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50139, Italy. .,Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, 50139, Italy.
| |
Collapse
|
18
|
Tubular Cell Cycle Response upon AKI: Revising Old and New Paradigms to Identify Novel Targets for CKD Prevention. Int J Mol Sci 2021; 22:ijms222011093. [PMID: 34681750 PMCID: PMC8537394 DOI: 10.3390/ijms222011093] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023] Open
Abstract
Acute kidney injury (AKI) is characterized by a rapid deterioration of kidney function, representing a global healthcare concern. In addition, AKI survivors frequently develop chronic kidney disease (CKD), contributing to a substantial proportion of disease burden globally. Yet, over the past 30 years, the burden of CKD has not declined to the same extent as many other important non-communicable diseases, implying a substantial deficit in the understanding of the disease progression. The assumption that the kidney response to AKI is based on a high proliferative potential of proximal tubular cells (PTC) caused a critical confounding factor, which has led to a limited development of strategies to prevent AKI and halt progression toward CKD. In this review, we discuss the latest findings on multiple mechanisms of response related to cell cycle behavior of PTC upon AKI, with a specific focus on their biological relevance. Collectively, we aim to (1) provide a new perspective on interpreting cell cycle progression of PTC in response to damage and (2) discuss how this knowledge can be used to choose the right therapeutic window of treatment for preserving kidney function while avoiding CKD progression.
Collapse
|
19
|
Losick VP, Duhaime LG. The endocycle restores tissue tension in the Drosophila abdomen post wound repair. Cell Rep 2021; 37:109827. [PMID: 34644579 PMCID: PMC8567445 DOI: 10.1016/j.celrep.2021.109827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/16/2021] [Accepted: 09/22/2021] [Indexed: 01/04/2023] Open
Abstract
Polyploidy frequently arises in response to injury, aging, and disease. Despite its prevalence, major gaps exist in our understanding of how polyploid cells alter tissue function. In the adult Drosophila epithelium, wound healing is dependent on the generation of multinucleated polyploid cells resulting in a permanent change in the epithelial architecture. Here, we study how the wound-induced polyploid cells affect tissue function by altering epithelial mechanics. The mechanosensor nonmuscle myosin II is activated and upregulated in wound-induced polyploid cells and persists after healing completes. Polyploidy enhances relative epithelial tension, which is dependent on the endocycle and not cell fusion post injury. Remarkably, the enhanced epithelial tension mimics the relative tension of the lateral muscle fibers, which are permanently severed by the injury. As a result, we found that the wound-induced polyploid cells remodel the epithelium to maintain fly abdominal movements, which may help compensate for lost tissue tension. Losick and Duhaime show that the generation of polyploid cells by the endocycle induces myosin expression resulting in enhanced epithelial tension after wound repair. This change in epithelial mechanics appears to compensate for the permanent loss of muscle fibers, which is necessary for efficient abdominal bending in the fruit fly.
Collapse
Affiliation(s)
- Vicki P Losick
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA.
| | - Levi G Duhaime
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA
| |
Collapse
|
20
|
Bailey EC, Kobielski S, Park J, Losick VP. Polyploidy in Tissue Repair and Regeneration. Cold Spring Harb Perspect Biol 2021; 13:a040881. [PMID: 34187807 PMCID: PMC8485745 DOI: 10.1101/cshperspect.a040881] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Polyploidy is defined as a cell with three or more whole genome sets and enables cell growth across the kingdoms of life. Studies in model organisms have revealed that polyploid cell growth can be required for optimal tissue repair and regeneration. In mammals, polyploid cell growth contributes to repair of many tissues, including the liver, heart, kidney, bladder, and eye, and similar strategies have been identified in Drosophila and zebrafish tissues. This review discusses the heterogeneity and versatility of polyploidy in tissue repair and regeneration. Polyploidy has been shown to restore tissue mass and maintain organ size as well as protect against oncogenic insults and genotoxic stress. Polyploid cells can also serve as a reservoir for new diploid cells in regeneration. The numerous mechanisms to generate polyploid cells provide an unlimited resource for tissues to exploit to undergo repair or regeneration.
Collapse
Affiliation(s)
- Erin C Bailey
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467, USA
| | - Sara Kobielski
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467, USA
| | - John Park
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467, USA
| | - Vicki P Losick
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467, USA
| |
Collapse
|
21
|
Almeida Machado Costa C, Wang XF, Ellsworth C, Deng WM. Polyploidy in development and tumor models in Drosophila. Semin Cancer Biol 2021; 81:106-118. [PMID: 34562587 DOI: 10.1016/j.semcancer.2021.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/03/2021] [Accepted: 09/18/2021] [Indexed: 12/26/2022]
Abstract
Polyploidy, a cell status defined as more than two sets of genomic DNA, is a conserved strategy across species that can increase cell size and biosynthetic production, but the functional aspects of polyploidy are nuanced and vary across cell types. Throughout Drosophila developmental stages (embryo, larva, pupa and adult), polyploid cells are present in numerous organs and help orchestrate development while contributing to normal growth, well-being and homeostasis of the organism. Conversely, increasing evidence has shown that polyploid cells are prevalent in Drosophila tumors and play important roles in tumor growth and invasiveness. Here, we summarize the genes and pathways involved in polyploidy during normal and tumorigenic development, the mechanisms underlying polyploidization, and the functional aspects of polyploidy in development, homeostasis and tumorigenesis in the Drosophila model.
Collapse
Affiliation(s)
- Caique Almeida Machado Costa
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, United States
| | - Xian-Feng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, United States
| | - Calder Ellsworth
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, United States
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, United States.
| |
Collapse
|
22
|
Kirillova A, Han L, Liu H, Kühn B. Polyploid cardiomyocytes: implications for heart regeneration. Development 2021; 148:271050. [PMID: 34897388 DOI: 10.1242/dev.199401] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Terminally differentiated cells are generally thought to have arrived at their final form and function. Many terminally differentiated cell types are polyploid, i.e. they have multiple copies of the normally diploid genome. Mammalian heart muscle cells, termed cardiomyocytes, are one such example of polyploid cells. Terminally differentiated cardiomyocytes are bi- or multi-nucleated, or have polyploid nuclei. Recent mechanistic studies of polyploid cardiomyocytes indicate that they can limit cellular proliferation and, hence, heart regeneration. In this short Spotlight, we present the mechanisms generating bi- and multi-nucleated cardiomyocytes, and the mechanisms generating polyploid nuclei. Our aim is to develop hypotheses about how these mechanisms might relate to cardiomyocyte proliferation and cardiac regeneration. We also discuss how these new findings could be applied to advance cardiac regeneration research, and how they relate to studies of other polyploid cells, such as cancer cells.
Collapse
Affiliation(s)
- Anna Kirillova
- Medical Scientist Training Program, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA 15219, USA
| | - Lu Han
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Honghai Liu
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Bernhard Kühn
- Division of Cardiology, UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children's Hospital of Pittsburgh and Department of Pediatrics, 4401 Penn Ave, Pittsburgh, PA 15224, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
23
|
Nandakumar S, Rozich E, Buttitta L. Cell Cycle Re-entry in the Nervous System: From Polyploidy to Neurodegeneration. Front Cell Dev Biol 2021; 9:698661. [PMID: 34249947 PMCID: PMC8264763 DOI: 10.3389/fcell.2021.698661] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Terminally differentiated cells of the nervous system have long been considered to be in a stable non-cycling state and are often considered to be permanently in G0. Exit from the cell cycle during development is often coincident with the differentiation of neurons, and is critical for neuronal function. But what happens in long lived postmitotic tissues that accumulate cell damage or suffer cell loss during aging? In other contexts, cells that are normally non-dividing or postmitotic can or re-enter the cell cycle and begin replicating their DNA to facilitate cellular growth in response to cell loss. This leads to a state called polyploidy, where cells contain multiple copies of the genome. A growing body of literature from several vertebrate and invertebrate model organisms has shown that polyploidy in the nervous system may be more common than previously appreciated and occurs under normal physiological conditions. Moreover, it has been found that neuronal polyploidization can play a protective role when cells are challenged with DNA damage or oxidative stress. By contrast, work over the last two and a half decades has discovered a link between cell-cycle reentry in neurons and several neurodegenerative conditions. In this context, neuronal cell cycle re-entry is widely considered to be aberrant and deleterious to neuronal health. In this review, we highlight historical and emerging reports of polyploidy in the nervous systems of various vertebrate and invertebrate organisms. We discuss the potential functions of polyploidization in the nervous system, particularly in the context of long-lived cells and age-associated polyploidization. Finally, we attempt to reconcile the seemingly disparate associations of neuronal polyploidy with both neurodegeneration and neuroprotection.
Collapse
Affiliation(s)
| | | | - Laura Buttitta
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
24
|
Shapiro JA. What can evolutionary biology learn from cancer biology? PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 165:19-28. [PMID: 33930405 DOI: 10.1016/j.pbiomolbio.2021.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022]
Abstract
Detecting and treating cancer effectively involves understanding the disease as one of somatic cell and tumor macroevolution. That understanding is key to avoid triggering an adverse reaction to therapy that generates an untreatable and deadly tumor population. Macroevolution differs from microevolution by karyotype changes rather than isolated localized mutations being the major source of hereditary variation. Cancer cells display major multi-site chromosome rearrangements that appear to have arisen in many different cases abruptly in the history of tumor evolution. These genome restructuring events help explain the punctuated macroevolutionary changes that mark major transitions in cancer progression. At least two different nonrandom patterns of rapid multisite genome restructuring - chromothripsis ("chromosome shattering") and chromoplexy ("chromosome weaving") - are clearly distinct in their distribution within the genome and in the cell biology of the stress-induced processes responsible for their occurrence. These observations tell us that eukaryotic cells have the capacity to reorganize their genomes rapidly in response to calamity. Since chromothripsis and chromoplexy have been identified in the human germline and in other eukaryotes, they provide a model for organismal macroevolution in response to the kinds of stresses that lead to mass extinctions.
Collapse
Affiliation(s)
- James A Shapiro
- Department of Biochemistry and Molecular Biology, University of Chicago, United States.
| |
Collapse
|
25
|
How Chaotic Is Genome Chaos? Cancers (Basel) 2021; 13:cancers13061358. [PMID: 33802828 PMCID: PMC8002653 DOI: 10.3390/cancers13061358] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Cancer genomes can undergo major restructurings involving many chromosomal locations at key stages in tumor development. This restructuring process has been designated “genome chaos” by some authors. In order to examine how chaotic cancer genome restructuring may be, the cell and molecular processes for DNA restructuring are reviewed. Examination of the action of these processes in various cancers reveals a degree of specificity that indicates genome restructuring may be sufficiently reproducible to enable possible therapies that interrupt tumor progression to more lethal forms. Abstract Cancer genomes evolve in a punctuated manner during tumor evolution. Abrupt genome restructuring at key steps in this evolution has been called “genome chaos.” To answer whether widespread genome change is truly chaotic, this review (i) summarizes the limited number of cell and molecular systems that execute genome restructuring, (ii) describes the characteristic signatures of DNA changes that result from activity of those systems, and (iii) examines two cases where genome restructuring is determined to a significant degree by cell type or viral infection. The conclusion is that many restructured cancer genomes display sufficiently unchaotic signatures to identify the cellular systems responsible for major oncogenic transitions, thereby identifying possible targets for therapies to inhibit tumor progression to greater aggressiveness.
Collapse
|
26
|
Cell-cell fusions and cell-in-cell phenomena in healthy cells and cancer: Lessons from protists and invertebrates. Semin Cancer Biol 2021; 81:96-105. [PMID: 33713795 DOI: 10.1016/j.semcancer.2021.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/28/2021] [Accepted: 03/04/2021] [Indexed: 02/08/2023]
Abstract
Herein we analyze two special routes of the multinucleated cells' formation - the fusion of mononuclear cells and the formation of cell-in-cell structures - in the healthy tissues and in tumorigenesis. There are many theories of tumorigenesis based on the phenomenon of emergence of the hybrid cancer cells. We consider the phenomena, which are rarely mentioned in those theories: namely, cellularization of syncytium or coenocytes, and the reversible or irreversible somatogamy. The latter includes the short-term and the long-term vegetative (somatic) cells' fusions in the life cycles of unicellular organisms. The somatogamy and multinuclearity have repeatedly and independently emerged in various groups of unicellular eukaryotes. These phenomena are among dominant survival and biodiversity sustaining strategies in protists and we admit that they can likely play an analogous role in cancer cells.
Collapse
|
27
|
Besen-McNally R, Gjelsvik KJ, Losick VP. Wound-induced polyploidization is dependent on Integrin-Yki signaling. Biol Open 2021; 10:bio.055996. [PMID: 33355119 PMCID: PMC7860123 DOI: 10.1242/bio.055996] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A key step in tissue repair is to replace lost or damaged cells. This occurs via two strategies: restoring cell number through proliferation or increasing cell size through polyploidization. Studies in Drosophila and vertebrates have demonstrated that polyploid cells arise in adult tissues, at least in part, to promote tissue repair and restore tissue mass. However, the signals that cause polyploid cells to form in response to injury remain poorly understood. In the adult Drosophila epithelium, wound-induced polyploid cells are generated by both cell fusion and endoreplication, resulting in a giant polyploid syncytium. Here, we identify the integrin focal adhesion complex as an activator of wound-induced polyploidization. Both integrin and focal adhesion kinase are upregulated in the wound-induced polyploid cells and are required for Yorkie-induced endoreplication and cell fusion. As a result, wound healing is perturbed when focal adhesion genes are knocked down. These findings show that conserved focal adhesion signaling is required to initiate wound-induced polyploid cell growth.
Collapse
Affiliation(s)
- Rose Besen-McNally
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 0×4469, USA
| | - Kayla J Gjelsvik
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 0×4469, USA.,Kathryn W. Davis Center for Regenerative Biology and Aging, MDI Biological Laboratory, Bar Harbor, ME, 04609, USA
| | - Vicki P Losick
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| |
Collapse
|
28
|
Communal living: the role of polyploidy and syncytia in tissue biology. Chromosome Res 2021; 29:245-260. [PMID: 34075512 PMCID: PMC8169410 DOI: 10.1007/s10577-021-09664-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/10/2021] [Accepted: 05/16/2021] [Indexed: 01/22/2023]
Abstract
Multicellular organisms are composed of tissues with diverse cell sizes. Whether a tissue primarily consists of numerous, small cells as opposed to fewer, large cells can impact tissue development and function. The addition of nuclear genome copies within a common cytoplasm is a recurring strategy to manipulate cellular size within a tissue. Cells with more than two genomes can exist transiently, such as in developing germlines or embryos, or can be part of mature somatic tissues. Such nuclear collectives span multiple levels of organization, from mononuclear or binuclear polyploid cells to highly multinucleate structures known as syncytia. Here, we review the diversity of polyploid and syncytial tissues found throughout nature. We summarize current literature concerning tissue construction through syncytia and/or polyploidy and speculate why one or both strategies are advantageous.
Collapse
|
29
|
Nakamura M, Verboon JM, Allen TE, Abreu-Blanco MT, Liu R, Dominguez ANM, Delrow JJ, Parkhurst SM. Autocrine insulin pathway signaling regulates actin dynamics in cell wound repair. PLoS Genet 2020; 16:e1009186. [PMID: 33306674 PMCID: PMC7758051 DOI: 10.1371/journal.pgen.1009186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/23/2020] [Accepted: 10/09/2020] [Indexed: 01/13/2023] Open
Abstract
Cells are exposed to frequent mechanical and/or chemical stressors that can compromise the integrity of the plasma membrane and underlying cortical cytoskeleton. The molecular mechanisms driving the immediate repair response launched to restore the cell cortex and circumvent cell death are largely unknown. Using microarrays and drug-inhibition studies to assess gene expression, we find that initiation of cell wound repair in the Drosophila model is dependent on translation, whereas transcription is required for subsequent steps. We identified 253 genes whose expression is up-regulated (80) or down-regulated (173) in response to laser wounding. A subset of these genes were validated using RNAi knockdowns and exhibit aberrant actomyosin ring assembly and/or actin remodeling defects. Strikingly, we find that the canonical insulin signaling pathway controls actin dynamics through the actin regulators Girdin and Chickadee (profilin), and its disruption leads to abnormal wound repair. Our results provide new insight for understanding how cell wound repair proceeds in healthy individuals and those with diseases involving wound healing deficiencies.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Jeffrey M. Verboon
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Tessa E. Allen
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Maria Teresa Abreu-Blanco
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Raymond Liu
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Andrew N. M. Dominguez
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Jeffrey J. Delrow
- Genomics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Susan M. Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| |
Collapse
|
30
|
Nandakumar S, Grushko O, Buttitta LA. Polyploidy in the adult Drosophila brain. eLife 2020; 9:e54385. [PMID: 32840209 PMCID: PMC7447450 DOI: 10.7554/elife.54385] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
Long-lived cells such as terminally differentiated postmitotic neurons and glia must cope with the accumulation of damage over the course of an animal's lifespan. How long-lived cells deal with ageing-related damage is poorly understood. Here we show that polyploid cells accumulate in the adult fly brain and that polyploidy protects against DNA damage-induced cell death. Multiple types of neurons and glia that are diploid at eclosion, become polyploid in the adult Drosophila brain. The optic lobes exhibit the highest levels of polyploidy, associated with an elevated DNA damage response in this brain region. Inducing oxidative stress or exogenous DNA damage leads to an earlier onset of polyploidy, and polyploid cells in the adult brain are more resistant to DNA damage-induced cell death than diploid cells. Our results suggest polyploidy may serve a protective role for neurons and glia in adult Drosophila melanogaster brains.
Collapse
Affiliation(s)
- Shyama Nandakumar
- Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Olga Grushko
- Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Laura A Buttitta
- Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| |
Collapse
|
31
|
Bailey EC, Dehn AS, Gjelsvik KJ, Besen-McNally R, Losick VP. A Drosophila Model to Study Wound-induced Polyploidization. J Vis Exp 2020. [PMID: 32597839 DOI: 10.3791/61252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Polyploidy is a frequent phenomenon whose impact on organismal health and disease is still poorly understood. A cell is defined as polyploid if it contains more than the diploid copy of its chromosomes, which is a result of endoreplication or cell fusion. In tissue repair, wound-induced polyploidization (WIP) has been found to be a conserved healing strategy from fruit flies to vertebrates. WIP has several advantages over cell proliferation, including resistance to oncogenic growth and genotoxic stress. The challenge has been to identify why polyploid cells arise and how these unique cells function. Provided is a detailed protocol to study WIP in the adult fruit fly epithelium where polyploid cells are generated within 2 days after a puncture wound. Taking advantage of D. melanogaster's extensive genetic tool kit, the genes required to initiate and regulate WIP, including Myc, have begun to be identified. Continued studies using this method can reveal how other genetic and physiological variables including sex, diet, and age regulate and influence WIP's function.
Collapse
Affiliation(s)
| | | | - Kayla J Gjelsvik
- Graduate School of Biomedical Sciences and Engineering and Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, University of Maine
| | | | | |
Collapse
|
32
|
Ren D, Song J, Ni M, Kang L, Guo W. Regulatory Mechanisms of Cell Polyploidy in Insects. Front Cell Dev Biol 2020; 8:361. [PMID: 32548115 PMCID: PMC7272692 DOI: 10.3389/fcell.2020.00361] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
Polyploidy cells undergo the endocycle to generate DNA amplification without cell division and have important biological functions in growth, development, reproduction, immune response, nutrient support, and conferring resistance to DNA damage in animals. In this paper, we have specially summarized current research progresses in the regulatory mechanisms of cell polyploidy in insects. First, insect hormones including juvenile hormone and 20-hydroxyecdysone regulate the endocycle of variant cells in diverse insect species. Second, cells skip mitotic division in response to developmental programming and conditional stimuli such as wound healing, regeneration, and aging. Third, the reported regulatory pathways of mitotic to endocycle switch (MES), including Notch, Hippo, and JNK signaling pathways, are summarized and constructed into genetic network. Thus, we think that the studies in crosstalk of hormones and their effects on canonical pathways will shed light on the mechanism of cell polyploidy and elucidate the evolutionary adaptions of MES through diverse insect species.
Collapse
Affiliation(s)
- Dani Ren
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Juan Song
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Ming Ni
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, Hebei University, Baoding, China
| | - Le Kang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, Hebei University, Baoding, China
| | - Wei Guo
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
33
|
Abstract
Drosophila melanogaster has historically been a workhorse model organism for studying developmental biology. In addition, Drosophila is an excellent model for studying how damaged tissues and organs can regenerate. Recently, new precision approaches that enable both highly targeted injury and genetic manipulation have accelerated progress in this field. Here, we highlight these techniques and review examples of recently discovered mechanisms that regulate regeneration in Drosophila larval and adult tissues. We also discuss how, by applying these powerful approaches, studies of Drosophila can continue to guide the future of regeneration research.
Collapse
Affiliation(s)
- Donald T Fox
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Erez Cohen
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Rachel Smith-Bolton
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
34
|
Cohen E, Sawyer JK, Peterson NG, Dow JAT, Fox DT. Physiology, Development, and Disease Modeling in the Drosophila Excretory System. Genetics 2020; 214:235-264. [PMID: 32029579 PMCID: PMC7017010 DOI: 10.1534/genetics.119.302289] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
The insect excretory system contains two organ systems acting in concert: the Malpighian tubules and the hindgut perform essential roles in excretion and ionic and osmotic homeostasis. For over 350 years, these two organs have fascinated biologists as a model of organ structure and function. As part of a recent surge in interest, research on the Malpighian tubules and hindgut of Drosophila have uncovered important paradigms of organ physiology and development. Further, many human disease processes can be modeled in these organs. Here, focusing on discoveries in the past 10 years, we provide an overview of the anatomy and physiology of the Drosophila excretory system. We describe the major developmental events that build these organs during embryogenesis, remodel them during metamorphosis, and repair them following injury. Finally, we highlight the use of the Malpighian tubules and hindgut as accessible models of human disease biology. The Malpighian tubule is a particularly excellent model to study rapid fluid transport, neuroendocrine control of renal function, and modeling of numerous human renal conditions such as kidney stones, while the hindgut provides an outstanding model for processes such as the role of cell chirality in development, nonstem cell-based injury repair, cancer-promoting processes, and communication between the intestine and nervous system.
Collapse
Affiliation(s)
| | - Jessica K Sawyer
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, and
| | | | - Julian A T Dow
- Institute of Molecular, Cell, and Systems Biology, University of Glasgow, G12 8QQ, United Kingdom
| | - Donald T Fox
- Department of Cell Biology and
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, and
| |
Collapse
|
35
|
Defoe DM, Rao H, Harris DJ, Moore PD, Brocher J, Harrison TA. A non-canonical role for p27Kip1 in restricting proliferation of corneal endothelial cells during development. PLoS One 2020; 15:e0226725. [PMID: 31929545 PMCID: PMC6957298 DOI: 10.1371/journal.pone.0226725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/30/2019] [Indexed: 12/04/2022] Open
Abstract
The cell cycle regulator p27Kip1 is a critical factor controlling cell number in many lineages. While its anti-proliferative effects are well-established, the extent to which this is a result of its function as a cyclin-dependent kinase (CDK) inhibitor or through other known molecular interactions is not clear. To genetically dissect its role in the developing corneal endothelium, we examined mice harboring two loss-of-function alleles, a null allele (p27−) that abrogates all protein function and a knockin allele (p27CK−) that targets only its interaction with cyclins and CDKs. Whole-animal mutants, in which all cells are either homozygous knockout or knockin, exhibit identical proliferative increases (~0.6-fold) compared with wild-type tissues. On the other hand, use of mosaic analysis with double markers (MADM) to produce infrequently-occurring clones of wild-type and mutant cells within the same tissue environment uncovers a roughly three- and six-fold expansion of individual p27CK−/CK− and p27−/− cells, respectively. Mosaicism also reveals distinct migration phenotypes, with p27−/− cells being highly restricted to their site of production and p27CK−/CK− cells more widely scattered within the endothelium. Using a density-based clustering algorithm to quantify dispersal of MADM-generated clones, a four-fold difference in aggregation is seen between the two types of mutant cells. Overall, our analysis reveals that, in developing mouse corneal endothelium, p27 regulates cell number by acting cell autonomously, both through its interactions with cyclins and CDKs and through a cyclin-CDK-independent mechanism(s). Combined with its parallel influence on cell motility, it constitutes a potent multi-functional effector mechanism with major impact on tissue organization.
Collapse
Affiliation(s)
- Dennis M. Defoe
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- * E-mail:
| | - Huiying Rao
- Department of Ophthalmology, Fujian Provincial Hospital, Fujian, Fuzhou, Peoples Republic of China
| | - David J. Harris
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Preston D. Moore
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- Graduate Biomedical Research Program, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | | | - Theresa A. Harrison
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| |
Collapse
|
36
|
Lee CW, Kwon YC, Lee Y, Park MY, Choe KM. cdc37 is essential for JNK pathway activation and wound closure in Drosophila. Mol Biol Cell 2019; 30:2651-2658. [PMID: 31483695 PMCID: PMC6761768 DOI: 10.1091/mbc.e18-12-0822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Wound closure in the Drosophila larval epidermis mainly involves nonproliferative, endocyling epithelial cells. Consequently, it is largely mediated by cell growth and migration. We discovered that both cell growth and migration in Drosophila require the cochaperone-encoding gene cdc37. Larvae lacking cdc37 in the epidermis failed to close wounds, and the cells of the epidermis failed to change cell shape and polarize. Likewise, wound-induced cell growth was significantly reduced, and correlated with a reduction in the size of the cell nucleus. The c-Jun N-terminal kinase (JNK) pathway, which is essential for wound closure, was not typically activated in injured cdc37 knockdown larvae. In addition, JNK, Hep, Mkk4, and Tak1 protein levels were reduced, consistent with previous reports showing that Cdc37 is important for the stability of various client kinases. Protein levels of the integrin β subunit and its wound-induced protein expression were also reduced, reflecting the disruption of JNK activation, which is crucial for expression of integrin β during wound closure. These results are consistent with a role of Cdc37 in maintaining the stability of the JNK pathway kinases, thus mediating cell growth and migration during Drosophila wound healing.
Collapse
Affiliation(s)
- Chan-Wool Lee
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Young-Chang Kwon
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Youngbin Lee
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Min-Yoon Park
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Kwang-Min Choe
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| |
Collapse
|
37
|
Grendler J, Lowgren S, Mills M, Losick VP. Wound-induced polyploidization is driven by Myc and supports tissue repair in the presence of DNA damage. Development 2019; 146:dev173005. [PMID: 31315896 PMCID: PMC6703715 DOI: 10.1242/dev.173005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 07/05/2019] [Indexed: 12/19/2022]
Abstract
Tissue repair usually requires either polyploid cell growth or cell division, but the molecular mechanism promoting polyploidy and limiting cell division remains poorly understood. Here, we find that injury to the adult Drosophila epithelium causes cells to enter the endocycle through the activation of Yorkie-dependent genes (Myc and E2f1). Myc is even sufficient to induce the endocycle in the uninjured post-mitotic epithelium. As result, epithelial cells enter S phase but mitosis is blocked by inhibition of mitotic gene expression. The mitotic cell cycle program can be activated by simultaneously expressing the Cdc25-like phosphatase String (stg), while genetically depleting APC/C E3 ligase fizzy-related (fzr). However, forcing cells to undergo mitosis is detrimental to wound repair as the adult fly epithelium accumulates DNA damage, and mitotic errors ensue when cells are forced to proliferate. In conclusion, we find that wound-induced polyploidization enables tissue repair when cell division is not a viable option.
Collapse
Affiliation(s)
- Janelle Grendler
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, 159 Old Bar Harbor Rd, Bar Harbor, ME 04609, USA
| | - Sara Lowgren
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, 159 Old Bar Harbor Rd, Bar Harbor, ME 04609, USA
| | - Monique Mills
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, 159 Old Bar Harbor Rd, Bar Harbor, ME 04609, USA
| | - Vicki P Losick
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, 159 Old Bar Harbor Rd, Bar Harbor, ME 04609, USA
| |
Collapse
|
38
|
Rotelli MD, Policastro RA, Bolling AM, Killion AW, Weinberg AJ, Dixon MJ, Zentner GE, Walczak CE, Lilly MA, Calvi BR. A Cyclin A-Myb-MuvB-Aurora B network regulates the choice between mitotic cycles and polyploid endoreplication cycles. PLoS Genet 2019; 15:e1008253. [PMID: 31291240 PMCID: PMC6645565 DOI: 10.1371/journal.pgen.1008253] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 07/22/2019] [Accepted: 06/18/2019] [Indexed: 12/30/2022] Open
Abstract
Endoreplication is a cell cycle variant that entails cell growth and periodic genome duplication without cell division, and results in large, polyploid cells. Cells switch from mitotic cycles to endoreplication cycles during development, and also in response to conditional stimuli during wound healing, regeneration, aging, and cancer. In this study, we use integrated approaches in Drosophila to determine how mitotic cycles are remodeled into endoreplication cycles, and how similar this remodeling is between induced and developmental endoreplicating cells (iECs and devECs). Our evidence suggests that Cyclin A / CDK directly activates the Myb-MuvB (MMB) complex to induce transcription of a battery of genes required for mitosis, and that repression of CDK activity dampens this MMB mitotic transcriptome to promote endoreplication in both iECs and devECs. iECs and devECs differed, however, in that devECs had reduced expression of E2F1-dependent genes that function in S phase, whereas repression of the MMB transcriptome in iECs was sufficient to induce endoreplication without a reduction in S phase gene expression. Among the MMB regulated genes, knockdown of AurB protein and other subunits of the chromosomal passenger complex (CPC) induced endoreplication, as did knockdown of CPC-regulated cytokinetic, but not kinetochore, proteins. Together, our results indicate that the status of a CycA-Myb-MuvB-AurB network determines the decision to commit to mitosis or switch to endoreplication in both iECs and devECs, and suggest that regulation of different steps of this network may explain the known diversity of polyploid cycle types in development and disease.
Collapse
Affiliation(s)
- Michael D. Rotelli
- Department of Biology. Indiana University, Bloomington, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Robert A. Policastro
- Department of Biology. Indiana University, Bloomington, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Anna M. Bolling
- Department of Biology. Indiana University, Bloomington, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Andrew W. Killion
- Department of Biology. Indiana University, Bloomington, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Abraham J. Weinberg
- Department of Biology. Indiana University, Bloomington, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Michael J. Dixon
- Department of Biology. Indiana University, Bloomington, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Gabriel E. Zentner
- Department of Biology. Indiana University, Bloomington, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| | - Claire E. Walczak
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
- Indiana University School of Medicine, Bloomington, Indiana, United States of America
| | - Mary A. Lilly
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Brian R. Calvi
- Department of Biology. Indiana University, Bloomington, Indiana, United States of America
- Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
- Indiana University School of Medicine, Bloomington, Indiana, United States of America
| |
Collapse
|
39
|
Erban T, Sopko B, Kadlikova K, Talacko P, Harant K. Varroa destructor parasitism has a greater effect on proteome changes than the deformed wing virus and activates TGF-β signaling pathways. Sci Rep 2019; 9:9400. [PMID: 31253851 PMCID: PMC6599063 DOI: 10.1038/s41598-019-45764-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 06/10/2019] [Indexed: 02/07/2023] Open
Abstract
Honeybee workers undergo metamorphosis in capped cells for approximately 13 days before adult emergence. During the same period, Varroa mites prick the defenseless host many times. We sought to identify proteome differences between emerging Varroa-parasitized and parasite-free honeybees showing the presence or absence of clinical signs of deformed wing virus (DWV) in the capped cells. A label-free proteomic analysis utilizing nanoLC coupled with an Orbitrap Fusion Tribrid mass spectrometer provided a quantitative comparison of 2316 protein hits. Redundancy analysis (RDA) showed that the combination of Varroa parasitism and DWV clinical signs caused proteome changes that occurred in the same direction as those of Varroa alone and were approximately two-fold higher. Furthermore, proteome changes associated with DWV signs alone were positioned above Varroa in the RDA. Multiple markers indicate that Varroa activates TGF-β-induced pathways to suppress wound healing and the immune response and that the collective action of stressors intensifies these effects. Furthermore, we indicate JAK/STAT hyperactivation, p53-BCL-6 feedback loop disruption, Wnt pathway activation, Wnt/Hippo crosstalk disruption, and NF-κB and JAK/STAT signaling conflict in the Varroa–honeybee–DWV interaction. These results illustrate the higher effect of Varroa than of DWV at the time of emergence. Markers for future research are provided.
Collapse
Affiliation(s)
- Tomas Erban
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne, CZ-161 06, Czechia.
| | - Bruno Sopko
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne, CZ-161 06, Czechia
| | - Klara Kadlikova
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne, CZ-161 06, Czechia.,Department of Plant Protection, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences, Prague 6-Suchdol, CZ-165 00, Czechia
| | - Pavel Talacko
- Proteomics Core Facility, Faculty of Science, Charles University, BIOCEV, Prumyslova 595, Vestec, CZ-25242, Czechia
| | - Karel Harant
- Proteomics Core Facility, Faculty of Science, Charles University, BIOCEV, Prumyslova 595, Vestec, CZ-25242, Czechia
| |
Collapse
|
40
|
Lazzeri E, Angelotti ML, Conte C, Anders HJ, Romagnani P. Surviving Acute Organ Failure: Cell Polyploidization and Progenitor Proliferation. Trends Mol Med 2019; 25:366-381. [PMID: 30935780 DOI: 10.1016/j.molmed.2019.02.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 12/20/2022]
Abstract
In acute organ failure, rapid compensation of function loss assures survival. Dedifferentiation and/or proliferation of surviving parenchymal cells could imply a transient (and potentially fatal) impairment of residual functional performance. However, evolution has selected two flexible life-saving mechanisms acting synergistically on organ function recovery. Sustaining residual performance is possible when the remnant differentiated parenchymal cells avoid cell division, but increase function by undergoing hypertrophy via endoreplication, leading to polyploid cells. In addition, tissue progenitors, representing a subset of less-differentiated and/or self-renewing parenchymal cells completing cytokinesis, proliferate and differentiate to regenerate lost parenchymal cells. Here, we review the evolving evidence on polyploidization and progenitor-driven regeneration in acute liver, heart, and kidney failure with evolutionary advantages and trade-offs in organ repair.
Collapse
Affiliation(s)
- Elena Lazzeri
- Department of Biological and Experimental Medical Science 'Mario Serio', Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE)
| | - Maria Lucia Angelotti
- Department of Biological and Experimental Medical Science 'Mario Serio', Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE)
| | - Carolina Conte
- Department of Biological and Experimental Medical Science 'Mario Serio', Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE)
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Paola Romagnani
- Department of Biological and Experimental Medical Science 'Mario Serio', Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE); Meyer Children's Hospital, Florence, Italy. http://www.twitter.com/PRomagnani
| |
Collapse
|
41
|
Nuclear Scaling Is Coordinated among Individual Nuclei in Multinucleated Muscle Fibers. Dev Cell 2019; 49:48-62.e3. [PMID: 30905770 DOI: 10.1016/j.devcel.2019.02.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/28/2018] [Accepted: 02/22/2019] [Indexed: 12/22/2022]
Abstract
Optimal cell performance depends on cell size and the appropriate relative size, i.e., scaling, of the nucleus. How nuclear scaling is regulated and contributes to cell function is poorly understood, especially in skeletal muscle fibers, which are among the largest cells, containing hundreds of nuclei. Here, we present a Drosophila in vivo system to analyze nuclear scaling in whole multinucleated muscle fibers, genetically manipulate individual components, and assess muscle function. Despite precise global coordination, we find that individual nuclei within a myofiber establish different local scaling relationships by adjusting their size and synthetic activity in correlation with positional or spatial cues. While myonuclei exhibit compensatory potential, even minor changes in global nuclear size scaling correlate with reduced muscle function. Our study provides the first comprehensive approach to unraveling the intrinsic regulation of size in multinucleated muscle fibers. These insights to muscle cell biology will accelerate the development of interventions for muscle diseases.
Collapse
|
42
|
Gjelsvik KJ, Besen-McNally R, Losick VP. Solving the Polyploid Mystery in Health and Disease. Trends Genet 2019; 35:6-14. [PMID: 30470486 PMCID: PMC6457904 DOI: 10.1016/j.tig.2018.10.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/12/2018] [Accepted: 10/22/2018] [Indexed: 01/12/2023]
Abstract
Polyploidy (the more than doubling of a cell's genome) frequently arises during organogenesis, tissue repair, and age-associated diseases. Despite its prevalence, major gaps exist in how polyploid cells emerge and affect tissue function. Studies have begun to elucidate the signals required for polyploid cell growth as well as the advantages and disadvantages of polyploidy in health and disease. This review highlights the recent advances on the role and regulation of polyploidy in Drosophila and vertebrate models. The newly discovered versatility of polyploid cells has the potential to provide alternative strategies to promote tissue growth and repair, while limiting disease and dysfunction.
Collapse
Affiliation(s)
- K J Gjelsvik
- MDI Biological Laboratory, 159 Old Bar Harbor Road, Bar Harbor, ME 04609, USA
| | - R Besen-McNally
- MDI Biological Laboratory, 159 Old Bar Harbor Road, Bar Harbor, ME 04609, USA
| | - V P Losick
- MDI Biological Laboratory, 159 Old Bar Harbor Road, Bar Harbor, ME 04609, USA.
| |
Collapse
|
43
|
Cytoplasmic localization of GRHL3 upon epidermal differentiation triggers cell shape change for epithelial morphogenesis. Nat Commun 2018; 9:4059. [PMID: 30283008 PMCID: PMC6170465 DOI: 10.1038/s41467-018-06171-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 08/16/2018] [Indexed: 11/08/2022] Open
Abstract
Epithelial cell shape change is a pivotal driving force for morphogenesis of complex three-dimensional architecture. However, molecular mechanisms triggering shape changes of epithelial cells in the course of growth and differentiation have not been entirely elucidated. Grhl3 plays a crucial role as a downstream transcription factor of Wnt/β-catenin in epidermal differentiation. Here, we show Grhl3 induced large, mature epidermal cells, enriched with actomyosin networks, from embryoid bodies in vitro. Such epidermal cells were apparently formed by the simultaneous activation of canonical and non-canonical Wnt signaling pathways. A nuclear transcription factor, GRHL3 is localized in the cytoplasm and cell membrane during epidermal differentiation. Subsequently, such extranuclear GRHL3 is essential for the membrane-associated expression of VANGL2 and CELSR1. Cytoplasmic GRHL3, thereby, allows epidermal cells to acquire mechanical properties for changes in epithelial cell shape. Thus, we propose that cytoplasmic localization of GRHL3 upon epidermal differentiation directly triggers epithelial morphogenesis.
Collapse
|
44
|
Park SH, Lee CW, Lee JH, Park JY, Roshandell M, Brennan CA, Choe KM. Requirement for and polarized localization of integrin proteins during Drosophila wound closure. Mol Biol Cell 2018; 29:2137-2147. [PMID: 29995573 PMCID: PMC6249799 DOI: 10.1091/mbc.e17-11-0635] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 06/19/2018] [Accepted: 07/05/2018] [Indexed: 01/06/2023] Open
Abstract
Wound reepithelialization is an evolutionarily conserved process in which skin cells migrate as sheets to heal the breach and is critical to prevent infection but impaired in chronic wounds. Integrin heterodimers mediate attachment between epithelia and underlying extracellular matrix and also act in large signaling complexes. The complexity of the mammalian wound environment and evident redundancy among integrins has impeded determination of their specific contributions to reepithelialization. Taking advantage of the genetic tools and smaller number of integrins in Drosophila, we undertook a systematic in vivo analysis of integrin requirements in the reepithelialization of skin wounds in the larva. We identify αPS2-βPS and αPS3-βPS as the crucial integrin dimers and talin as the only integrin adhesion component required for reepithelialization. The integrins rapidly accumulate in a JNK-dependent manner in a few rows of cells surrounding a wound. Intriguingly, the integrins localize to the distal margin in these cells, instead of the frontal or lamellipodial distribution expected for proteins providing traction and recruit nonmuscle myosin II to the same location. These findings indicate that signaling roles of integrins may be important for epithelial polarization around wounds and lay the groundwork for using Drosophila to better understand integrin contributions to reepithelialization.
Collapse
Affiliation(s)
- Si-Hyoung Park
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Chan-wool Lee
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Ji-Hyun Lee
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Jin Young Park
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Mobina Roshandell
- Department of Biological Science, California State University, Fullerton, Fullerton, CA 92831
| | - Catherine A. Brennan
- Department of Biological Science, California State University, Fullerton, Fullerton, CA 92831
| | - Kwang-Min Choe
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| |
Collapse
|
45
|
Cohen E, Allen SR, Sawyer JK, Fox DT. Fizzy-Related dictates A cell cycle switch during organ repair and tissue growth responses in the Drosophila hindgut. eLife 2018; 7:e38327. [PMID: 30117808 PMCID: PMC6130973 DOI: 10.7554/elife.38327] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
Ploidy-increasing cell cycles drive tissue growth in many developing organs. Such cycles, including endocycles, are increasingly appreciated to drive tissue growth following injury or activated growth signaling in mature organs. In these organs, the regulation and distinct roles of different cell cycles remains unclear. Here, we uncover a programmed switch between cell cycles in the Drosophila hindgut pylorus. Using an acute injury model, we identify mitosis as the response in larval pyloric cells, whereas endocycles occur in adult pyloric cells. By developing a novel genetic method, DEMISE (Dual-Expression-Method-for-Induced-Site-specific-Eradication), we show the cell cycle regulator Fizzy-related dictates the decision between mitosis and endocycles. After injury, both cycles accurately restore tissue mass and genome content. However, in response to sustained growth signaling, only endocycles preserve epithelial architecture. Our data reveal distinct cell cycle programming in response to similar stimuli in mature vs. developmental states and reveal a tissue-protective role of endocycles.
Collapse
Affiliation(s)
- Erez Cohen
- Department of Cell BiologyDuke University School of MedicineDurhamUnited States
| | - Scott R Allen
- Department of Cell BiologyDuke University School of MedicineDurhamUnited States
| | - Jessica K Sawyer
- Department of Pharmacology & Cancer BiologyDuke University School of MedicineDurhamUnited States
| | - Donald T Fox
- Department of Cell BiologyDuke University School of MedicineDurhamUnited States
- Department of Pharmacology & Cancer BiologyDuke University School of MedicineDurhamUnited States
- Regeneration Next InitiativeDuke University School of MedicineDurhamUnited States
| |
Collapse
|
46
|
Abstract
Polyploid cells, which contain multiple copies of the typically diploid genome, are widespread in plants and animals. Polyploidization can be developmentally programmed or stress induced, and arises from either cell-cell fusion or a process known as endoreplication, in which cells replicate their DNA but either fail to complete cytokinesis or to progress through M phase entirely. Polyploidization offers cells several potential fitness benefits, including the ability to increase cell size and biomass production without disrupting cell and tissue structure, and allowing improved cell longevity through higher tolerance to genomic stress and apoptotic signals. Accordingly, recent studies have uncovered crucial roles for polyploidization in compensatory cell growth during tissue regeneration in the heart, liver, epidermis and intestine. Here, we review current knowledge of the molecular pathways that generate polyploidy and discuss how polyploidization is used in tissue repair and regeneration.
Collapse
Affiliation(s)
| | - Bruce A Edgar
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| |
Collapse
|
47
|
Vazquez-Martin A, Anatskaya OV, Giuliani A, Erenpreisa J, Huang S, Salmina K, Inashkina I, Huna A, Nikolsky NN, Vinogradov AE. Somatic polyploidy is associated with the upregulation of c-MYC interacting genes and EMT-like signature. Oncotarget 2018; 7:75235-75260. [PMID: 27655693 PMCID: PMC5342737 DOI: 10.18632/oncotarget.12118] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/05/2016] [Indexed: 12/30/2022] Open
Abstract
The dependence of cancer on overexpressed c-MYC and its predisposition for polyploidy represents a double puzzle. We address this conundrum by cross-species transcription analysis of c-MYC interacting genes in polyploid vs. diploid tissues and cells, including human vs. mouse heart, mouse vs. human liver and purified 4n vs. 2n mouse decidua cells. Gene-by-gene transcriptome comparison and principal component analysis indicated that c-MYC interactants are significantly overrepresented among ploidy-associated genes. Protein interaction networks and gene module analysis revealed that the most upregulated genes relate to growth, stress response, proliferation, stemness and unicellularity, as well as to the pathways of cancer supported by MAPK and RAS coordinated pathways. A surprising feature was the up-regulation of epithelial-mesenchymal transition (EMT) modules embodied by the N-cadherin pathway and EMT regulators from SNAIL and TWIST families. Metabolic pathway analysis also revealed the EMT-linked features, such as global proteome remodeling, oxidative stress, DNA repair and Warburg-like energy metabolism. Genes associated with apoptosis, immunity, energy demand and tumour suppression were mostly down-regulated. Noteworthy, despite the association between polyploidy and ample features of cancer, polyploidy does not trigger it. Possibly it occurs because normal polyploidy does not go that far in embryonalisation and linked genome destabilisation. In general, the analysis of polyploid transcriptome explained the evolutionary relation of c-MYC and polyploidy to cancer.
Collapse
Affiliation(s)
| | - Olga V Anatskaya
- Institute of Cytology, St-Petersburg, Russian Federation, Russia
| | | | | | - Sui Huang
- Systems Biology Institute, Seattle, USA
| | | | - Inna Inashkina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Anda Huna
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | |
Collapse
|
48
|
Spradling AC. Polytene Chromosome Structure and Somatic Genome Instability. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2017; 82:293-304. [PMID: 29167281 DOI: 10.1101/sqb.2017.82.033670] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Polytene chromosomes have for 80 years provided the highest resolution view of interphase genome structure in an animal cell nucleus. These chromosomes represent the normal genomic state of nearly all Drosophila larval and many adult cells, and a better understanding of their striking banded structure has been sought for decades. A more recently appreciated characteristic of Drosophila polytene cells is somatic genome instability caused by unfinished replication (UR). Repair of stalled forks generates enough deletions in polytene salivary gland cells to alter 10%-90% of the DNA strands within more than 100 UR regions comprising 20% of the euchromatic genome. We accurately map UR regions and show that most approximate large polytene bands, indicating that replication forks frequently stall near band boundaries in late S phase. Chromosome conformation capture has recently identified dense topologically associated domains (TADs) in many genomes and most UR bands are similar or slightly smaller than a cognate Drosophila TAD. We argue that bands serve the evolutionarily ancient function of coordinating genome replication with local gene activity. We also discuss the relatively recent evolution of polyteny and somatic instability in Diptera and propose that these processes helped propel the amazing success of two-winged flies in becoming the most ecologically diverse insect group, with 200 times the number of species as mammals.
Collapse
Affiliation(s)
- Allan C Spradling
- Department of Embryology, Howard Hughes Medical Institute, Carnegie Institution for Science, Baltimore, Maryland 21218
| |
Collapse
|
49
|
Cellular, ultrastructural and molecular analyses of epidermal cell development in the planarian Schmidtea mediterranea. Dev Biol 2017; 433:357-373. [PMID: 29100657 DOI: 10.1016/j.ydbio.2017.08.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/23/2017] [Accepted: 08/26/2017] [Indexed: 12/20/2022]
Abstract
The epidermis is essential for animal survival, providing both a protective barrier and cellular sensor to external environments. The generally conserved embryonic origin of the epidermis, but the broad morphological and functional diversity of this organ across animals is puzzling. We define the transcriptional regulators underlying epidermal lineage differentiation in the planarian Schmidtea mediterranea, an invertebrate organism that, unlike fruitflies and nematodes, continuously replaces its epidermal cells. We find that Smed-p53, Sox and Pax transcription factors are essential regulators of epidermal homeostasis, and act cooperatively to regulate genes associated with early epidermal precursor cell differentiation, including a tandemly arrayed novel gene family (prog) of secreted proteins. Additionally, we report on the discovery of distinct and previously undescribed secreted organelles whose production is dependent on the transcriptional activity of soxP-3, and which we term Hyman vesicles.
Collapse
|
50
|
Tsai CR, Anderson AE, Burra S, Jo J, Galko MJ. Yorkie regulates epidermal wound healing in Drosophila larvae independently of cell proliferation and apoptosis. Dev Biol 2017; 427:61-71. [PMID: 28514643 DOI: 10.1016/j.ydbio.2017.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/01/2017] [Accepted: 05/09/2017] [Indexed: 12/19/2022]
Abstract
Yorkie (Yki), the transcriptional co-activator of the Hippo signaling pathway, has well-characterized roles in balancing apoptosis and cell division during organ growth control. Yki is also required in diverse tissue regenerative contexts. In most cases this requirement reflects its well-characterized roles in balancing apoptosis and cell division. Whether Yki has repair functions outside of the control of cell proliferation, death, and growth is not clear. Here we show that Yki and Scalloped (Sd) are required for epidermal wound closure in the Drosophila larval epidermis. Using a GFP-tagged Yki transgene we show that Yki transiently translocates to some epidermal nuclei upon wounding. Genetic analysis strongly suggests that Yki interacts with the known wound healing pathway, Jun N-terminal kinase (JNK), but not with Platelet Derived Growth Factor/Vascular-Endothelial Growth Factor receptor (Pvr). Yki likely acts downstream of or parallel to JNK signaling and does not appear to regulate either proliferation or apoptosis in the larval epidermis during wound repair. Analysis of actin structures after wounding suggests that Yki and Sd promote wound closure through actin regulation. In sum, we found that Yki regulates an epithelial tissue repair process independently of its previously documented roles in balancing proliferation and apoptosis.
Collapse
Affiliation(s)
- Chang-Ru Tsai
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Aimee E Anderson
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sirisha Burra
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juyeon Jo
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Genes and Development Graduate Program, Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, TX 77030, USA
| | - Michael J Galko
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Genes and Development Graduate Program, Graduate School of Biomedical Sciences, The University of Texas M.D. Anderson Cancer Center, TX 77030, USA.
| |
Collapse
|