201
|
Epigenetics and Shared Molecular Processes in the Regeneration of Complex Structures. Stem Cells Int 2015; 2016:6947395. [PMID: 26681954 PMCID: PMC4670690 DOI: 10.1155/2016/6947395] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 05/30/2015] [Indexed: 12/27/2022] Open
Abstract
The ability to regenerate complex structures is broadly represented in both plant and animal kingdoms. Although regenerative abilities vary significantly amongst metazoans, cumulative studies have identified cellular events that are broadly observed during regenerative events. For example, structural damage is recognized and wound healing initiated upon injury, which is followed by programmed cell death in the vicinity of damaged tissue and a burst in proliferation of progenitor cells. Sustained proliferation and localization of progenitor cells to site of injury give rise to an assembly of differentiating cells known as the regeneration blastema, which fosters the development of new tissue. Finally, preexisting tissue rearranges and integrates with newly differentiated cells to restore proportionality and function. While heterogeneity exists in the basic processes displayed during regenerative events in different species—most notably the cellular source contributing to formation of new tissue—activation of conserved molecular pathways is imperative for proper regulation of cells during regeneration. Perhaps the most fundamental of such molecular processes entails chromatin rearrangements, which prime large changes in gene expression required for differentiation and/or dedifferentiation of progenitor cells. This review provides an overview of known contributions to regenerative processes by noncoding RNAs and chromatin-modifying enzymes involved in epigenetic regulation.
Collapse
|
202
|
Phagocytosis of apoptotic cells in homeostasis. Nat Immunol 2015; 16:907-17. [PMID: 26287597 DOI: 10.1038/ni.3253] [Citation(s) in RCA: 626] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/17/2015] [Indexed: 02/07/2023]
Abstract
Human bodies collectively turn over about 200 billion to 300 billion cells every day. Such turnover is an integral part of embryonic and postnatal development, as well as routine tissue homeostasis. This process involves the induction of programmed cell death in specific cells within the tissues and the specific recognition and removal of dying cells by a clearance 'crew' composed of professional, non-professional and specialized phagocytes. In the past few years, considerable progress has been made in identifying many features of apoptotic cell clearance. Some of these new observations challenge the way dying cells themselves are viewed, as well as how healthy cells interact with and respond to dying cells. Here we focus on the homeostatic removal of apoptotic cells in tissues.
Collapse
|
203
|
Petrenko V, Mihhailova J, Salmon P, Kiss JZ. Apoptotic neurons induce proliferative responses of progenitor cells in the postnatal neocortex. Exp Neurol 2015; 273:126-37. [PMID: 26291762 DOI: 10.1016/j.expneurol.2015.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 07/13/2015] [Accepted: 08/11/2015] [Indexed: 12/29/2022]
Abstract
Apoptotic cell death is the leading cause of neuronal loss after neonatal brain injury. Little is known about the intrinsic capacity of the immature cerebral cortex for replacing dead cells. Here we test the hypothesis that neuronal apoptosis is able to trigger compensatory proliferation in surrounding cells. In order to establish a "pure" apoptotic cell death model and to avoid the confounding effects of broken blood-brain barrier and inflammatory reactions, we used a diphtheria toxin (DT) and diphtheria toxin receptor (DTR) system to induce ablation of layer IV neurons in the rodent somatosensory cortex during the early postnatal period. We found that DT-triggered apoptosis is a slowly progressing event lasting about for 7 days. While dying cells expressed the morphological features of apoptosis, we could not detect immunoreactivity for activated caspase-3 in these cells. Microglia activation and proliferation represented the earliest cellular responses to apoptotic cell death. In addition, we found that induced apoptosis triggered a massive proliferation of undifferentiated progenitor cell pool including Sox2 as well as NG2 cells. The default differentiation pattern of proliferating progenitors appears to be the glial phenotype; we could not find evidence for newly generated neurons in response to apoptotic neuronal death. These results suggest that mitotically active progenitor populations are intrinsically capable to contribute to the repair process of injured cortical tissue and may represent a potential target for neuronal replacement strategies.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Department of Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Jevgenia Mihhailova
- Department of Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Patrick Salmon
- Department of Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | - Jozsef Z Kiss
- Department of Neurosciences, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland.
| |
Collapse
|
204
|
Buzgariu W, Al Haddad S, Tomczyk S, Wenger Y, Galliot B. Multi-functionality and plasticity characterize epithelial cells in Hydra. Tissue Barriers 2015; 3:e1068908. [PMID: 26716072 PMCID: PMC4681288 DOI: 10.1080/21688370.2015.1068908] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/23/2015] [Accepted: 06/27/2015] [Indexed: 01/09/2023] Open
Abstract
Epithelial sheets, a synapomorphy of all metazoans but porifers, are present as 2 layers in cnidarians, ectoderm and endoderm, joined at their basal side by an extra-cellular matrix named mesoglea. In the Hydra polyp, epithelial cells of the body column are unipotent stem cells that continuously self-renew and concomitantly express their epitheliomuscular features. These multifunctional contractile cells maintain homeostasis by providing a protective physical barrier, by digesting nutrients, by selecting a stable microbiota, and by rapidly closing wounds. In addition, epithelial cells are highly plastic, supporting the adaptation of Hydra to physiological and environmental changes, such as long starvation periods where survival relies on a highly dynamic autophagy flux. Epithelial cells also play key roles in developmental processes as evidenced by the organizer activity they develop to promote budding and regeneration. We propose here an integrative view of the homeostatic and developmental aspects of epithelial plasticity in Hydra.
Collapse
Affiliation(s)
- W Buzgariu
- Department of Genetics and Evolution; Institute of Genetics and Genomics in Geneva (IGe3); Faculty of Sciences; University of Geneva; Geneva, Switzerland
| | - S Al Haddad
- Department of Genetics and Evolution; Institute of Genetics and Genomics in Geneva (IGe3); Faculty of Sciences; University of Geneva; Geneva, Switzerland
| | - S Tomczyk
- Department of Genetics and Evolution; Institute of Genetics and Genomics in Geneva (IGe3); Faculty of Sciences; University of Geneva; Geneva, Switzerland
| | - Y Wenger
- Department of Genetics and Evolution; Institute of Genetics and Genomics in Geneva (IGe3); Faculty of Sciences; University of Geneva; Geneva, Switzerland
| | - B Galliot
- Department of Genetics and Evolution; Institute of Genetics and Genomics in Geneva (IGe3); Faculty of Sciences; University of Geneva; Geneva, Switzerland
| |
Collapse
|
205
|
Zhou N, Wang R, Zhang Y, Lei Z, Zhang X, Hu R, Li H, Mao Y, Wang X, Irwin DM, Niu G, Tan H. Staurosporine Induced Apoptosis May Activate Cancer Stem-Like Cells (CD44(+)/CD24(-)) in MCF-7 by Upregulating Mucin1 and EpCAM. J Cancer 2015; 6:1049-57. [PMID: 26366219 PMCID: PMC4565855 DOI: 10.7150/jca.12501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 07/27/2015] [Indexed: 12/31/2022] Open
Abstract
Malignant tumors recur after chemotherapy. A small population of cancer stem-like cells within tumors is now generally considered the prime source of the recurrence. To better understand how cancer stem-like cells induce relapse after fractionated chemotherapy, we examined changes in the CD44(+)/CD24(-) cancer stem-like cells population and behavior using the breast cancer cell line MCF-7. Our results show that apart from an increase in the CD44(+)/CD24(-) population, proliferation and clone formation, but not migration, were enhanced after recovery from apoptosis induced by two pulses of staurosporine (STS). The distribution of cells in the cell cycle differed between acutely induced apoptosis and fractionated chemotherapy. Sorted CD44(+)/CD24(-) stem-like cells from MCF-7 cells recovered from STS treatment possessed greater proliferation abilities. We also observed that mucin1 (MUC1) and Epithelial cell adhesion molecule (EpCAM) were up-regulated in abundance coincidently with proliferation and clone formation enhancement. Our findings suggest that fractionated chemotherapy induced apoptosis could stimulate cancer stem-like cell to behave with a stronger malignant property than cancer cells themselves and MUC1 and EpCAM are important factors involving in this process. By demonstrating changes in cancer stem cell during chemotherapy and identifying the crucial factors, we potentially can target them, to eradicate tumors and overcome cancer relapse.
Collapse
Affiliation(s)
- Na Zhou
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| | - Rong Wang
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| | - Yizhuang Zhang
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| | - Zhen Lei
- 2. Beijing N&N Genetech Company, Beijing, 100082, China
| | - Xuehui Zhang
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| | - Ruobi Hu
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| | - Hui Li
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| | - Yiqing Mao
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| | - Xi Wang
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| | - David M Irwin
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China; ; 3. Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Gang Niu
- 2. Beijing N&N Genetech Company, Beijing, 100082, China
| | - Huanran Tan
- 1. Department of Pharmacology, Peking University, Health Science Center, Beijing 100191, China
| |
Collapse
|
206
|
Wang H, Lööf S, Borg P, Nader GA, Blau HM, Simon A. Turning terminally differentiated skeletal muscle cells into regenerative progenitors. Nat Commun 2015; 6:7916. [PMID: 26243583 PMCID: PMC4765497 DOI: 10.1038/ncomms8916] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/24/2015] [Indexed: 01/09/2023] Open
Abstract
The ability to repeatedly regenerate limbs during the entire lifespan of an animal is restricted to certain salamander species among vertebrates. This ability involves dedifferentiation of post-mitotic cells into progenitors that in turn form new structures. A long-term enigma has been how injury leads to dedifferentiation. Here we show that skeletal muscle dedifferentiation during newt limb regeneration depends on a programmed cell death response by myofibres. We find that programmed cell death-induced muscle fragmentation produces a population of ‘undead' intermediate cells, which have the capacity to resume proliferation and contribute to muscle regeneration. We demonstrate the derivation of proliferating progeny from differentiated, multinucleated muscle cells by first inducing and subsequently intercepting a programmed cell death response. We conclude that cell survival may be manifested by the production of a dedifferentiated cell with broader potential and that the diversion of a programmed cell death response is an instrument to achieve dedifferentiation. Newts can regenerate amputated limbs via unknown mechanism involving dedifferentiation of cells in the stump into progenitors that contribute to the new appendages. Here the authors show that skeletal muscle dedifferentiation in regenerating newt limbs relies on a diverted programmed cell death response by myofibers.
Collapse
Affiliation(s)
- Heng Wang
- Department of Cell and Molecular Biology, Karolinska Institute, Solna, Sweden
| | - Sara Lööf
- Department of Cell and Molecular Biology, Karolinska Institute, Solna, Sweden
| | - Paula Borg
- Department of Cell and Molecular Biology, Karolinska Institute, Solna, Sweden
| | - Gustavo A Nader
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Stanford University, Stanford, California 94305, United States
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Solna, Sweden
| |
Collapse
|
207
|
Petersen HO, Höger SK, Looso M, Lengfeld T, Kuhn A, Warnken U, Nishimiya-Fujisawa C, Schnölzer M, Krüger M, Özbek S, Simakov O, Holstein TW. A Comprehensive Transcriptomic and Proteomic Analysis of Hydra Head Regeneration. Mol Biol Evol 2015; 32:1928-47. [PMID: 25841488 PMCID: PMC4833066 DOI: 10.1093/molbev/msv079] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The cnidarian freshwater polyp Hydra sp. exhibits an unparalleled regeneration capacity in the animal kingdom. Using an integrative transcriptomic and stable isotope labeling by amino acids in cell culture proteomic/phosphoproteomic approach, we studied stem cell-based regeneration in Hydra polyps. As major contributors to head regeneration, we identified diverse signaling pathways adopted for the regeneration response as well as enriched novel genes. Our global analysis reveals two distinct molecular cascades: an early injury response and a subsequent, signaling driven patterning of the regenerating tissue. A key factor of the initial injury response is a general stabilization of proteins and a net upregulation of transcripts, which is followed by a subsequent activation cascade of signaling molecules including Wnts and transforming growth factor (TGF) beta-related factors. We observed moderate overlap between the factors contributing to proteomic and transcriptomic responses suggesting a decoupled regulation between the transcriptional and translational levels. Our data also indicate that interstitial stem cells and their derivatives (e.g., neurons) have no major role in Hydra head regeneration. Remarkably, we found an enrichment of evolutionarily more recent genes in the early regeneration response, whereas conserved genes are more enriched in the late phase. In addition, genes specific to the early injury response were enriched in transposon insertions. Genetic dynamicity and taxon-specific factors might therefore play a hitherto underestimated role in Hydra regeneration.
Collapse
Affiliation(s)
- Hendrik O Petersen
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Stefanie K Höger
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Mario Looso
- Max Planck Institute (MPI) for Heart and Lung Research, Bad Nauheim, Germany
| | - Tobias Lengfeld
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Anne Kuhn
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Uwe Warnken
- Functional Proteome Analysis Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Chiemi Nishimiya-Fujisawa
- Okazaki Institute for Integrative Bioscience, National Institute for Basic Biology, Myodaiji, Okazaki, Japan
| | - Martina Schnölzer
- Functional Proteome Analysis Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcus Krüger
- Max Planck Institute (MPI) for Heart and Lung Research, Bad Nauheim, Germany CECAD, University of Cologne, Germany
| | - Suat Özbek
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Oleg Simakov
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany Molecular Genetics Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Thomas W Holstein
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
208
|
Bagherie-Lachidan M, Reginensi A, Pan Q, Zaveri HP, Scott DA, Blencowe BJ, Helmbacher F, McNeill H. Stromal Fat4 acts non-autonomously with Dchs1/2 to restrict the nephron progenitor pool. Development 2015; 142:2564-73. [PMID: 26116661 DOI: 10.1242/dev.122648] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 06/18/2015] [Indexed: 01/10/2023]
Abstract
Regulation of the balance between progenitor self-renewal and differentiation is crucial to development. In the mammalian kidney, reciprocal signalling between three lineages (stromal, mesenchymal and ureteric) ensures correct nephron progenitor self-renewal and differentiation. Loss of either the atypical cadherin FAT4 or its ligand Dachsous 1 (DCHS1) results in expansion of the mesenchymal nephron progenitor pool, called the condensing mesenchyme (CM). This has been proposed to be due to misregulation of the Hippo kinase pathway transcriptional co-activator YAP. Here, we use tissue-specific deletions to prove that FAT4 acts non-autonomously in the renal stroma to control nephron progenitors. We show that loss of Yap from the CM in Fat4-null mice does not reduce the expanded CM, indicating that FAT4 regulates the CM independently of YAP. Analysis of Six2(-/-);Fat4(-/-) double mutants demonstrates that excess progenitors in Fat4 mutants are dependent on Six2, a crucial regulator of nephron progenitor self-renewal. Electron microscopy reveals that cell organisation is disrupted in Fat4 mutants. Gene expression analysis demonstrates that the expression of Notch and FGF pathway components are altered in Fat4 mutants. Finally, we show that Dchs1, and its paralogue Dchs2, function in a partially redundant fashion to regulate the number of nephron progenitors. Our data support a model in which FAT4 in the stroma binds to DCHS1/2 in the mouse CM to restrict progenitor self-renewal.
Collapse
Affiliation(s)
- Mazdak Bagherie-Lachidan
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8 Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada M5G 1X5
| | - Antoine Reginensi
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada M5G 1X5
| | - Qun Pan
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| | - Hitisha P Zaveri
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Benjamin J Blencowe
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8 Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| | | | - Helen McNeill
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8 Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada M5G 1X5
| |
Collapse
|
209
|
Myc regulates programmed cell death and radial glia dedifferentiation after neural injury in an echinoderm. BMC DEVELOPMENTAL BIOLOGY 2015; 15:24. [PMID: 26025619 PMCID: PMC4448152 DOI: 10.1186/s12861-015-0071-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/01/2015] [Indexed: 11/30/2022]
Abstract
Background Adult echinoderms can completely regenerate major parts of their central nervous system even after severe injuries. Even though this capacity has long been known, the molecular mechanisms that drive fast and complete regeneration in these animals have remained uninvestigated. The major obstacle for understanding these molecular pathways has been the lack of functional genomic studies on regenerating adult echinoderms. Results Here, we employ RNA interference-mediated gene knockdown to characterize the role of Myc during the early (first 48 hours) post-injury response in the radial nerve cord of the sea cucumber Holothuria glaberrima. Our previous experiments identified Myc as the only pluripotency-associated factor, whose expression significantly increased in the wounded CNS. The specific function(s) of this gene, however, remained unknown. Here we demonstrate that knockdown of Myc inhibits dedifferentiation of radial glia and programmed cell death, the two most prominent cellular events that take place in the regenerating sea cucumber nervous system shortly after injury. Conclusions In this study, we show that Myc overexpression is required for proper dedifferentiation of radial glial cells and for triggering the programmed cell death in the vicinity of the injury. Myc is thus the first transcription factor, whose functional role has been experimentally established in echinoderm regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0071-z) contains supplementary material, which is available to authorized users.
Collapse
|
210
|
Simón R, Aparicio R, Housden BE, Bray S, Busturia A. Drosophila p53 controls Notch expression and balances apoptosis and proliferation. Apoptosis 2015; 19:1430-43. [PMID: 24858703 DOI: 10.1007/s10495-014-1000-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A balance between cell proliferation and apoptosis is important for normal development and tissue homeostasis. Under stress conditions, the conserved tumor suppressor and transcription factor Dp53 induces apoptosis to contribute to the maintenance of homeostasis. However, in some cases Dp53-induced apoptosis results in the proliferation of surrounding non-apoptotic cells. To gain insight into the Dp53 function in the control of apoptosis and proliferation, we studied the interaction between the Drosophila Dp53 and Notch genes. We present evidence that simultaneous reduction of Dp53 and Notch function synergistically increases the wing phenotype of Notch heterozygous mutant flies. Further, we found that a Notch cis-regulatory element is responsive to loss and gain of Dp53 function and that over-expression of Dp53 up-regulates Notch mRNA and protein expression. These findings suggest not only that Dp53 and Notch act together to control wing development but also indicate that Dp53 transcriptionally regulates Notch expression. Moreover, using Notch gain and loss of function mutations we examined the relevance of Dp53 and Notch interactions in the process of Dp53-apoptosis induced proliferation. Results show that proliferation induced by Dp53 over-expression is dependent on Notch, thus identifying Notch as a new player in Dp53-induced proliferation. Interestingly, we found that Dp53-induced Notch activation and proliferation occurs even under conditions where apoptosis was inhibited. Our findings highlight the conservation between flies and vertebrates of the Dp53 and Notch cross-talk and suggest that Dp53 has a dual role regulating cell death and proliferation gene networks to control the homeostatic balance between apoptosis and proliferation.
Collapse
Affiliation(s)
- Rocío Simón
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, c) Nicolás Cabrera 1, 28049, Madrid, Spain
| | | | | | | | | |
Collapse
|
211
|
Abstract
The canonical role of p53 in preserving genome integrity and limiting carcinogenesis has been well established. In the presence of acute DNA-damage, oncogene deregulation and other forms of cellular stress, p53 orchestrates a myriad of pleiotropic processes to repair cellular damages and maintain homeostasis. Beside these well-studied functions of p53, recent studies in Drosophila have unraveled intriguing roles of Dmp53 in promoting cell division in apoptosis-induced proliferation, enhancing fitness and proliferation of the winner cell in cell competition and coordinating growth at the organ and organismal level in the presence of stress. In this review, we describe these new functions of Dmp53 and discuss their relevance in the context of carcinogenesis.
Collapse
Affiliation(s)
- Bertrand Mollereau
- Laboratory of Molecular Biology of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France,
| | | |
Collapse
|
212
|
Live to die another way: modes of programmed cell death and the signals emanating from dying cells. Nat Rev Mol Cell Biol 2015; 16:329-44. [PMID: 25991373 DOI: 10.1038/nrm3999] [Citation(s) in RCA: 469] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
All life ends in death, but perhaps one of life's grander ironies is that it also depends on death. Cell-intrinsic suicide pathways, termed programmed cell death (PCD), are crucial for animal development, tissue homeostasis and pathogenesis. Originally, PCD was almost synonymous with apoptosis; recently, however, alternative mechanisms of PCD have been reported. Here, we provide an overview of several distinct PCD mechanisms, namely apoptosis, autophagy and necroptosis. In addition, we discuss the complex signals that emanate from dying cells, which can either trigger regeneration or instruct additional killing. Further advances in understanding the physiological roles of the various mechanisms of cell death and their associated signals will be important to selectively manipulate PCD for therapeutic purposes.
Collapse
|
213
|
Bradshaw B, Thompson K, Frank U. Distinct mechanisms underlie oral vs aboral regeneration in the cnidarian Hydractinia echinata. eLife 2015; 4:e05506. [PMID: 25884246 PMCID: PMC4421858 DOI: 10.7554/elife.05506] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 04/16/2015] [Indexed: 12/12/2022] Open
Abstract
Cnidarians possess remarkable powers of regeneration, but the cellular and molecular mechanisms underlying this capability are unclear. Studying the hydrozoan Hydractinia echinata we show that a burst of stem cell proliferation occurs following decapitation, forming a blastema at the oral pole within 24 hr. This process is necessary for head regeneration. Knocking down Piwi1, Vasa, Pl10 or Ncol1 expressed by blastema cells inhibited regeneration but not blastema formation. EdU pulse-chase experiments and in vivo tracking of individual transgenic Piwi1+ stem cells showed that the cellular source for blastema formation is migration of stem cells from a remote area. Surprisingly, no blastema developed at the aboral pole after stolon removal. Instead, polyps transformed into stolons and then budded polyps. Hence, distinct mechanisms act to regenerate different body parts in Hydractinia. This model, where stem cell behavior can be monitored in vivo at single cell resolution, offers new insights for regenerative biology. DOI:http://dx.doi.org/10.7554/eLife.05506.001 Although all animals are capable of regenerating damaged tissue to some extent, a few—including jellyfish, coral, and their relatives—are able to regenerate entire lost body parts. Closely related species may have very different regeneration capabilities. This has led some researchers to propose that higher animals, such as mammals, still possess the ancient genes that allow entire body parts to regenerate, but that somehow the genes have been disabled during their evolution. Studying animals that can regenerate large parts of their bodies may therefore help scientists understand what prevents others, including humans, from doing so. An animal that is particularly useful for studies into regeneration is called Hydractinia echinata. These tiny marine animals make their homes on the shells of hermit crabs. They are small, transparent and stay fixed to one spot, making it easy for scientists to grow them in the laboratory and closely observe what is going on when they regenerate. Bradshaw et al. genetically engineered Hydractinia individuals to produce a fluorescent protein in their stem cells; these cells have the ability to become one of several kinds of mature cell, and often help to repair and grow tissues. This allowed the stem cells to be tracked using a microscope. When the head of Hydractinia was cut off, stem cells in the animals' mid body section migrated to the end where the head used to be and multiplied. These stem cells then created a bud (known as a blastema) that developed into a new, fully functional head within two days, allowing the animals to capture prey. Reducing the activity of certain stem cell genes prevented the new head from growing, but the bud still formed. Next, Bradshaw et al. removed a structure from the opposite end of the animal, called the stolon, which normally helps Hydractinia attach to hermit crabs shells. Stolons regenerated in a completely different way to heads. No bud formed. Instead, the remainder of the animal's body, which included the head and the body column, gradually transformed into a stolon rather than regenerating this structure, and only then grew a new body column and head. Therefore, different tissues in the same animal can regenerate in different ways. Understanding the ‘tricks’ used by animals like Hydractinia to regenerate may help translate these abilities to regenerative medicine. DOI:http://dx.doi.org/10.7554/eLife.05506.002
Collapse
Affiliation(s)
- Brian Bradshaw
- School of Natural Sciences and Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Kerry Thompson
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, National University of Ireland, Galway, Ireland
| | - Uri Frank
- School of Natural Sciences and Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| |
Collapse
|
214
|
Labi V, Erlacher M. How cell death shapes cancer. Cell Death Dis 2015; 6:e1675. [PMID: 25741600 PMCID: PMC4385913 DOI: 10.1038/cddis.2015.20] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 12/28/2014] [Accepted: 01/02/2015] [Indexed: 12/20/2022]
Abstract
Apoptosis has been established as a mechanism of anti-cancer defense. Members of the BCL-2 family are critical mediators of apoptotic cell death in health and disease, often found to be deregulated in cancer and believed to lead to the survival of malignant clones. However, over the years, a number of studies pointed out that a model in which cell death resistance unambiguously acts as a barrier against malignant disease might be too simple. This is based on paradoxical observations made in tumor patients as well as mouse models indicating that apoptosis can indeed drive tumor formation, at least under certain circumstances. One possible explanation for this phenomenon is that apoptosis can promote proliferation critically needed to compensate for cell loss, for example, upon therapy, and to restore tissue homeostasis. However, this, at the same time, can promote tumor development by allowing expansion of selected clones. Usually, tissue resident stem/progenitor cells are a major source for repopulation, some of them potentially carrying (age-, injury- or therapy-induced) genetic aberrations deleterious for the host. Thereby, apoptosis might drive genomic instability by facilitating the emergence of pathologic clones during phases of proliferation and subsequent replication stress-associated DNA damage. Tumorigenesis initiated by repeated cell attrition and repopulation, as confirmed in different genetic models, has parallels in human cancers, exemplified in therapy-induced secondary malignancies and myelodysplastic syndromes in patients with congenital bone marrow failure syndromes. Here, we aim to review evidence in support of the oncogenic role of stress-induced apoptosis.
Collapse
Affiliation(s)
- V Labi
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin 13125, Germany
- Immune Regulation and Cancer, Max-Delbrück-Center for Molecular Medicine, Robert Rössle-Strasse 10, 13125 Berlin, Germany. Tel: +49 30 9406 3462; Fax: +49 30 9406 2390; E-mail:
| | - M Erlacher
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center of Freiburg, Freiburg 79106, Germany
- Freiburg Institute for Advanced Studies, University of Freiburg, Freiburg 79104, Germany
| |
Collapse
|
215
|
|
216
|
Li Q, Yang H, Zhong TP. Regeneration across metazoan phylogeny: lessons from model organisms. J Genet Genomics 2015; 42:57-70. [PMID: 25697100 DOI: 10.1016/j.jgg.2014.12.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/18/2014] [Accepted: 12/21/2014] [Indexed: 01/09/2023]
Abstract
Comprehending the diversity of the regenerative potential across metazoan phylogeny represents a fundamental challenge in biology. Invertebrates like Hydra and planarians exhibit amazing feats of regeneration, in which an entire organism can be restored from minute body segments. Vertebrates like teleost fish and amphibians can also regrow large sections of the body. While this regenerative capacity is greatly attenuated in mammals, there are portions of major organs that remain regenerative. Regardless of the extent, there are common basic strategies to regeneration, including activation of adult stem cells and proliferation of differentiated cells. Here, we discuss the cellular features and molecular mechanisms that are involved in regeneration in different model organisms, including Hydra, planarians, zebrafish and newts as well as in several mammalian organs.
Collapse
Affiliation(s)
- Qiao Li
- State Key Laboratory of Genetic Engineering, Department of Genetics, Fudan University School of Life Science, Shanghai 200433, China
| | - Hao Yang
- State Key Laboratory of Genetic Engineering, Department of Genetics, Fudan University School of Life Science, Shanghai 200433, China
| | - Tao P Zhong
- State Key Laboratory of Genetic Engineering, Department of Genetics, Fudan University School of Life Science, Shanghai 200433, China; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
217
|
Castellana D, Paus R, Perez-Moreno M. Macrophages contribute to the cyclic activation of adult hair follicle stem cells. PLoS Biol 2014; 12:e1002002. [PMID: 25536657 PMCID: PMC4275176 DOI: 10.1371/journal.pbio.1002002] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 10/10/2014] [Indexed: 12/17/2022] Open
Abstract
Castellana, Paus, and Perez-Moreno discover that skin resident macrophages signal to skin stem cells via Wnt ligands to activate the hair follicle life cycle. Skin epithelial stem cells operate within a complex signaling milieu that orchestrates their lifetime regenerative properties. The question of whether and how immune cells impact on these stem cells within their niche is not well understood. Here we show that skin-resident macrophages decrease in number because of apoptosis before the onset of epithelial hair follicle stem cell activation during the murine hair cycle. This process is linked to distinct gene expression, including Wnt transcription. Interestingly, by mimicking this event through the selective induction of macrophage apoptosis in early telogen, we identify a novel involvement of macrophages in stem cell activation in vivo. Importantly, the macrophage-specific pharmacological inhibition of Wnt production delays hair follicle growth. Thus, perifollicular macrophages contribute to the activation of skin epithelial stem cells as a novel, additional cue that regulates their regenerative activity. This finding may have translational implications for skin repair, inflammatory skin diseases and cancer. The cyclic life of hair follicles consists of recurring phases of growth, decay, and rest. Previous studies have identified signals that prompt a new phase of hair growth through the activation of resting hair follicle stem cells (HF-SCs). In addition to these signals, recent findings have shown that cues arising from the neighboring skin environment, in which hair follicles dwell, also participate in controlling hair follicle growth. Here we show that skin resident macrophages surround and signal to resting HF-SCs, regulating their entry into a new phase of hair follicle growth. This process involves the death and activation of a fraction of resident macrophages— resulting in Wnt ligand release —that in turn activate HF-SCs. These findings reveal additional mechanisms controlling endogenous stem cell pools that are likely to be relevant for modulating stem cell regenerative capabilities. The results provide new insights that may have implications for the development of technologies with potential applications in regeneration, aging, and cancer.
Collapse
Affiliation(s)
- Donatello Castellana
- Epithelial Cell Biology Group, BBVA Foundation-CNIO Cancer Cell Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ralf Paus
- Institute of Inflammation and Repair, University of Manchester, Manchester, United Kingdom
- Department of Dermatology, University of Münster, Münster, Germany
| | - Mirna Perez-Moreno
- Epithelial Cell Biology Group, BBVA Foundation-CNIO Cancer Cell Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- * E-mail:
| |
Collapse
|
218
|
Shalini S, Dorstyn L, Dawar S, Kumar S. Old, new and emerging functions of caspases. Cell Death Differ 2014; 22:526-39. [PMID: 25526085 DOI: 10.1038/cdd.2014.216] [Citation(s) in RCA: 930] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 11/13/2014] [Accepted: 11/19/2014] [Indexed: 12/26/2022] Open
Abstract
Caspases are proteases with a well-defined role in apoptosis. However, increasing evidence indicates multiple functions of caspases outside apoptosis. Caspase-1 and caspase-11 have roles in inflammation and mediating inflammatory cell death by pyroptosis. Similarly, caspase-8 has dual role in cell death, mediating both receptor-mediated apoptosis and in its absence, necroptosis. Caspase-8 also functions in maintenance and homeostasis of the adult T-cell population. Caspase-3 has important roles in tissue differentiation, regeneration and neural development in ways that are distinct and do not involve any apoptotic activity. Several other caspases have demonstrated anti-tumor roles. Notable among them are caspase-2, -8 and -14. However, increased caspase-2 and -8 expression in certain types of tumor has also been linked to promoting tumorigenesis. Increased levels of caspase-3 in tumor cells causes apoptosis and secretion of paracrine factors that promotes compensatory proliferation in surrounding normal tissues, tumor cell repopulation and presents a barrier for effective therapeutic strategies. Besides this caspase-2 has emerged as a unique caspase with potential roles in maintaining genomic stability, metabolism, autophagy and aging. The present review focuses on some of these less studied and emerging functions of mammalian caspases.
Collapse
Affiliation(s)
- S Shalini
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia
| | - L Dorstyn
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia
| | - S Dawar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia
| | - S Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
219
|
Abstract
Adult stem cells face the challenge of maintaining tissue homeostasis by self-renewal while maintaining their proliferation potential over the lifetime of an organism. Continuous proliferation can cause genotoxic/metabolic stress that can compromise the genomic integrity of stem cells. To prevent stem cell exhaustion, highly proliferative adult tissues maintain a pool of quiescent stem cells that divide only in response to injury and thus remain protected from genotoxic stress. Hydra is a remarkable organism with highly proliferative stem cells and ability to regenerate at whole animal level. Intriguingly, hydra does not display consequences of high proliferation, such as senescence or tumour formation. In this study, we investigate if hydra harbours a pool of slow-cycling stem cells that could help prevent undesirable consequences of continuous proliferation. Hydra were pulsed with the thymidine analogue 5-ethynyl-2′-deoxyuridine (EdU) and then chased in the absence of EdU to monitor the presence of EdU-retaining cells. A significant number of undifferentiated cells of all three lineages in hydra retained EdU for about 8–10 cell cycles, indicating that these cells did not enter cell cycle. These label-retaining cells were resistant to hydroxyurea treatment and were predominantly in the G2 phase of cell cycle. Most significantly, similar to mammalian quiescent stem cells, these cells rapidly entered cell division during head regeneration. This study shows for the first time that, contrary to current beliefs, cells in hydra display heterogeneity in their cell cycle potential and the slow-cycling cells in this population enter cell cycle during head regeneration. These results suggest an early evolution of slow-cycling stem cells in multicellular animals.
Collapse
Affiliation(s)
- Niraimathi Govindasamy
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Campus, Bellary Road, Bangalore 560065, India
| | - Supriya Murthy
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Campus, Bellary Road, Bangalore 560065, India
| | - Yashoda Ghanekar
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Campus, Bellary Road, Bangalore 560065, India
| |
Collapse
|
220
|
Abstract
Recent studies in Drosophila, Hydra, planarians, zebrafish, mice, indicate that cell death can open paths to regeneration in adult animals. Indeed injury can induce cell death, itself triggering regeneration following an immediate instructive mechanism, whereby the dying cells release signals that induce cellular responses over short and/or long-range distances. Cell death can also provoke a sustained derepressing response through the elimination of cells that suppress regeneration in homeostatic conditions. Whether common properties support what we name "regenerative cell death," is currently unclear. As key parameters, we review here the injury proapoptotic signals, the signals released by the dying cells, the cellular responses, and their respective timing. ROS appears as a common signal triggering cell death through MAPK and/or JNK pathway activation. But the modes of ROS production vary, from a brief pulse upon wounding, to repeated waves as observed in the zebrafish fin where ROS supports two peaks of cell death. Indeed regenerative cell death can be restricted to the injury phase, as in Hydra, Drosophila, or biphasic, immediate, and delayed, as in planarians and zebrafish. The dying cells release in a caspase-dependent manner a variety of signaling molecules, cytokines, growth factors, but also prostaglandins or ATP as recorded in Drosophila, Hydra, mice, and zebrafish, respectively. Interestingly, the ROS-producing cells often resist to cell death, implying a complex paracrine mode of signaling to launch regeneration, involving ROS-producing cells, ROS-sensing cells that release signaling molecules upon caspase activation, and effector cells that respond to these signals by proliferating, migrating, and/or differentiating.
Collapse
Affiliation(s)
- Sophie Vriz
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris, France; University Paris-Diderot, Paris, France
| | - Silke Reiter
- Department of Genetics and Evolution, University of Geneva, Switzerland
| | - Brigitte Galliot
- Department of Genetics and Evolution, University of Geneva, Switzerland.
| |
Collapse
|
221
|
Clevers H, Loh KM, Nusse R. Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science 2014; 346:1248012. [PMID: 25278615 DOI: 10.1126/science.1248012] [Citation(s) in RCA: 1012] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cells fuel tissue development, renewal, and regeneration, and these activities are controlled by the local stem cell microenvironment, the "niche." Wnt signals emanating from the niche can act as self-renewal factors for stem cells in multiple mammalian tissues. Wnt proteins are lipid-modified, which constrains them to act as short-range cellular signals. The locality of Wnt signaling dictates that stem cells exiting the Wnt signaling domain differentiate, spatially delimiting the niche in certain tissues. In some instances, stem cells may act as or generate their own niche, enabling the self-organization of patterned tissues. In this Review, we discuss the various ways by which Wnt operates in stem cell control and, in doing so, identify an integral program for tissue renewal and regeneration.
Collapse
Affiliation(s)
- Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Centre Utrecht and CancerGenomics.nl, 3584CT Utrecht, Netherlands
| | - Kyle M Loh
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Roel Nusse
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
222
|
Rampon C, Gauron C, Meda F, Volovitch M, Vriz S. Adenosine enhances progenitor cell recruitment and nerve growth via its A2B receptor during adult fin regeneration. Purinergic Signal 2014; 10:595-602. [PMID: 25084769 DOI: 10.1007/s11302-014-9420-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/22/2014] [Indexed: 12/12/2022] Open
Abstract
A major issue in regenerative medicine is the control of progenitor cell mobilisation. Apoptosis has been reported as playing a role in cell plasticity, and it has been recently shown that apoptosis is necessary for organ and appendage regeneration. In this context, we explore its possible mode of action in progenitor cell recruitment during adult regeneration in zebrafish. Here, we show that apoptosis inhibition impairs blastema formation and nerve growth, both of which can be restored by exogenous adenosine acting through its A2B receptor. Moreover, adenosine increases the number of progenitor cells. Purinergic signalling is therefore an early and essential event in the pathway from lesion to blastema formation and provides new targets for manipulating cell plasticity in the adult.
Collapse
Affiliation(s)
- Christine Rampon
- Centre Interdisciplinaire de Recherche en biologie (CIRB), CNRS UMR 7241//INSERM U1050, Collège de France, 11, Place Marcelin Berthelot, 75231, Paris Cedex 05, France
| | | | | | | | | |
Collapse
|
223
|
The link between injury-induced stress and regenerative phenomena: A cellular and genetic synopsis. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:454-61. [PMID: 25088176 DOI: 10.1016/j.bbagrm.2014.07.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/25/2014] [Accepted: 07/28/2014] [Indexed: 12/24/2022]
Abstract
Injury is an inescapable phenomenon of life that affects animals at every physiological level. Yet, some animals respond to injury by rebuilding the damaged tissues whereas others are limited to scarring. Elucidating how a tissue insult from wounding leads to a regenerative response at the genetic level is essential to make regenerative advantages translational. It has become clear that animals with regenerative abilities recycle developmental programs after injury, reactivating genes that have lied dormant throughout adulthood. The question that is critical to our understanding of regeneration is how a specific set of developmentally important genes can be reactivated only after an acute tissue insult. Here, we review how injury-induced cellular stresses such as hypoxic, oxidative, and mechanical stress may contribute to the genomic and epigenetic changes that promote regeneration in animals. This article is part of a Special Issue entitled: Stress as a fundamental theme in cell plasticity.
Collapse
|
224
|
|
225
|
Almuedo-Castillo M, Crespo X, Seebeck F, Bartscherer K, Salò E, Adell T. JNK controls the onset of mitosis in planarian stem cells and triggers apoptotic cell death required for regeneration and remodeling. PLoS Genet 2014; 10:e1004400. [PMID: 24922054 PMCID: PMC4055413 DOI: 10.1371/journal.pgen.1004400] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 04/09/2014] [Indexed: 01/18/2023] Open
Abstract
Regeneration of lost tissues depends on the precise interpretation of molecular signals that control and coordinate the onset of proliferation, cellular differentiation and cell death. However, the nature of those molecular signals and the mechanisms that integrate the cellular responses remain largely unknown. The planarian flatworm is a unique model in which regeneration and tissue renewal can be comprehensively studied in vivo. The presence of a population of adult pluripotent stem cells combined with the ability to decode signaling after wounding enable planarians to regenerate a complete, correctly proportioned animal within a few days after any kind of amputation, and to adapt their size to nutritional changes without compromising functionality. Here, we demonstrate that the stress-activated c-jun-NH2-kinase (JNK) links wound-induced apoptosis to the stem cell response during planarian regeneration. We show that JNK modulates the expression of wound-related genes, triggers apoptosis and attenuates the onset of mitosis in stem cells specifically after tissue loss. Furthermore, in pre-existing body regions, JNK activity is required to establish a positive balance between cell death and stem cell proliferation to enable tissue renewal, remodeling and the maintenance of proportionality. During homeostatic degrowth, JNK RNAi blocks apoptosis, resulting in impaired organ remodeling and rescaling. Our findings indicate that JNK-dependent apoptotic cell death is crucial to coordinate tissue renewal and remodeling required to regenerate and to maintain a correctly proportioned animal. Hence, JNK might act as a hub, translating wound signals into apoptotic cell death, controlled stem cell proliferation and differentiation, all of which are required to coordinate regeneration and tissue renewal.
Collapse
Affiliation(s)
- María Almuedo-Castillo
- Department of Genetics and Institute of Biomedicine, University of Barcelona, Barcelona, Catalonia, Spain
| | - Xenia Crespo
- Department of Genetics and Institute of Biomedicine, University of Barcelona, Barcelona, Catalonia, Spain
| | - Florian Seebeck
- Max Planck Research Group Stem Cells and Regeneration, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Faculty of Medicine, University of Münster, Münster, Germany
| | - Kerstin Bartscherer
- Max Planck Research Group Stem Cells and Regeneration, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Faculty of Medicine, University of Münster, Münster, Germany
| | - Emili Salò
- Department of Genetics and Institute of Biomedicine, University of Barcelona, Barcelona, Catalonia, Spain
| | - Teresa Adell
- Department of Genetics and Institute of Biomedicine, University of Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
226
|
Zimmerman MA, Huang Q, Li F, Liu X, Li CY. Cell death-stimulated cell proliferation: a tissue regeneration mechanism usurped by tumors during radiotherapy. Semin Radiat Oncol 2014; 23:288-95. [PMID: 24012343 DOI: 10.1016/j.semradonc.2013.05.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The death of all the cancer cells in a tumor is the ultimate goal of cancer therapy. Therefore, much of the current effort in cancer research is focused on activating cellular machinery that facilitates cell death such as factors involved in causing apoptosis. However, recently, a number of studies point to some counterintuitive roles for apoptotic caspases in radiation therapy as well as in tissue regeneration. It appears that a major function of apoptotic caspases is to facilitate tissue regeneration and tumor cell repopulation during cancer therapy. Because tumor cell repopulation has been shown to be important for local tumor relapse, understanding the molecular mechanisms behind tumor repopulation would be important to enhance cancer radiotherapy. In this review, we discuss our current knowledge of these potentially paradigm-changing phenomena and mechanisms in various organisms and their implications on the development of novel cancer therapeutics and strategies.
Collapse
Affiliation(s)
- Mary A Zimmerman
- Department of Dermatology, Duke University Medical Center, Durham, NC
| | | | | | | | | |
Collapse
|
227
|
Connolly PF, Jäger R, Fearnhead HO. New roles for old enzymes: killer caspases as the engine of cell behavior changes. Front Physiol 2014; 5:149. [PMID: 24795644 PMCID: PMC3997007 DOI: 10.3389/fphys.2014.00149] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 03/28/2014] [Indexed: 12/31/2022] Open
Abstract
It has become increasingly clear that caspases, far from being merely cell death effectors, have a much wider range of functions within the cell. These functions are as diverse as signal transduction and cytoskeletal remodeling, and caspases are now known to have an essential role in cell proliferation, migration, and differentiation. There is also evidence that apoptotic cells themselves can direct the behavior of nearby cells through the caspase-dependent secretion of paracrine signaling factors. In some processes, including the differentiation of skeletal muscle myoblasts, both caspase activation in differentiating cells as well as signaling from apoptotic cells has been reported. Here, we review the non-apoptotic outcomes of caspase activity in a range of different model systems and attempt to integrate this knowledge.
Collapse
Affiliation(s)
- Patrick F Connolly
- Pharmacology and Therapeutics, National University of Ireland Galway Galway, Ireland
| | - Richard Jäger
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences Rheinbach, Germany
| | - Howard O Fearnhead
- Pharmacology and Therapeutics, National University of Ireland Galway Galway, Ireland
| |
Collapse
|
228
|
Buzgariu W, Crescenzi M, Galliot B. Robust G2 pausing of adult stem cells in Hydra. Differentiation 2014; 87:83-99. [PMID: 24703763 DOI: 10.1016/j.diff.2014.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 12/18/2022]
Abstract
Hydra is a freshwater hydrozoan polyp that constantly renews its two tissue layers thanks to three distinct stem cell populations that cannot replace each other, epithelial ectodermal, epithelial endodermal, and multipotent interstitial. These adult stem cells, located in the central body column, exhibit different cycling paces, slow for the epithelial, fast for the interstitial. To monitor the changes in cell cycling in Hydra, we established a fast and efficient flow cytometry procedure, which we validated by confirming previous findings, as the Nocodazole-induced reversible arrest of cell cycling in G2/M, and the mitogenic signal provided by feeding. Then to dissect the cycling and differentiation behaviors of the interstitial stem cells, we used the AEP_cnnos1 and AEP_Icy1 transgenic lines that constitutively express GFP in this lineage. For the epithelial lineages we used the sf-1 strain that rapidly eliminates the fast cycling cells upon heat-shock and progressively becomes epithelial. This study evidences similar cycling patterns for the interstitial and epithelial stem cells, which all alternate between the G2 and S-phases traversing a minimal G1-phase. We also found interstitial progenitors with a shorter G2 that pause in G1/G0. At the animal extremities, most cells no longer cycle, the epithelial cells terminally differentiate in G2 and the interstitial progenitors in G1/G0. At the apical pole ~80% cells are post-mitotic differentiated cells, reflecting the higher density of neurons and nematocytes in this region. We discuss how the robust G2 pausing of stem cells, maintained over weeks of starvation, may contribute to regeneration.
Collapse
Affiliation(s)
- Wanda Buzgariu
- Department of Genetics and Evolution, University of Geneva, Sciences III, 30 Quai Ernest Ansermet, CH-1211 Geneva 4, Switzerland
| | | | - Brigitte Galliot
- Department of Genetics and Evolution, University of Geneva, Sciences III, 30 Quai Ernest Ansermet, CH-1211 Geneva 4, Switzerland.
| |
Collapse
|
229
|
DuBuc TQ, Traylor-Knowles N, Martindale MQ. Initiating a regenerative response; cellular and molecular features of wound healing in the cnidarian Nematostella vectensis. BMC Biol 2014; 12:24. [PMID: 24670243 PMCID: PMC4229989 DOI: 10.1186/1741-7007-12-24] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/12/2014] [Indexed: 11/17/2022] Open
Abstract
Background Wound healing is the first stage of a series of cellular events that are necessary to initiate a regenerative response. Defective wound healing can block regeneration even in animals with a high regenerative capacity. Understanding how signals generated during wound healing promote regeneration of lost structures is highly important, considering that virtually all animals have the ability to heal but many lack the ability to regenerate missing structures. Cnidarians are the phylogenetic sister taxa to bilaterians and are highly regenerative animals. To gain a greater understanding of how early animals generate a regenerative response, we examined the cellular and molecular components involved during wound healing in the anthozoan cnidarian Nematostella vectensis. Results Pharmacological inhibition of extracellular signal-regulated kinases (ERK) signaling blocks regeneration and wound healing in Nematostella. We characterized early and late wound healing events through genome-wide microarray analysis, quantitative PCR, and in situ hybridization to identify potential wound healing targets. We identified a number of genes directly related to the wound healing response in other animals (metalloproteinases, growth factors, transcription factors) and suggest that glycoproteins (mucins and uromodulin) play a key role in early wound healing events. This study also identified a novel cnidarian-specific gene, for a thiamine biosynthesis enzyme (vitamin B synthesis), that may have been incorporated into the genome by lateral gene transfer from bacteria and now functions during wound healing. Lastly, we suggest that ERK signaling is a shared element of the early wound response for animals with a high regenerative capacity. Conclusions This research describes the temporal events involved during Nematostella wound healing, and provides a foundation for comparative analysis with other regenerative and non-regenerative species. We have shown that the same genes that heal puncture wounds are also activated after oral-aboral bisection, indicating a clear link with the initiation of regenerative healing. This study demonstrates the strength of using a forward approach (microarray) to characterize a developmental phenomenon (wound healing) at a phylogenetically important crossroad of animal evolution (cnidarian-bilaterian ancestor). Accumulation of data on the early wound healing events across numerous systems may provide clues as to why some animals have limited regenerative abilities.
Collapse
Affiliation(s)
| | | | - Mark Q Martindale
- University of Florida, Whitney Marine Laboratory, 9505 Oceanshore Boulevard, St, Augustine, FL 32080, USA.
| |
Collapse
|
230
|
Yamaguchi H, Maruyama T, Urade Y, Nagata S. Immunosuppression via adenosine receptor activation by adenosine monophosphate released from apoptotic cells. eLife 2014; 3:e02172. [PMID: 24668173 PMCID: PMC3963506 DOI: 10.7554/elife.02172] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Apoptosis is coupled with recruitment of macrophages for engulfment of dead cells, and with compensatory proliferation of neighboring cells. Yet, this death process is silent, and it does not cause inflammation. The molecular mechanisms underlying anti-inflammatory nature of the apoptotic process remains poorly understood. In this study, we found that the culture supernatant of apoptotic cells activated the macrophages to express anti-inflammatory genes such as Nr4a and Thbs1. A high level of AMP accumulated in the apoptotic cell supernatant in a Pannexin1-dependent manner. A nucleotidase inhibitor and A2a adenosine receptor antagonist inhibited the apoptotic supernatant-induced gene expression, suggesting AMP was metabolized to adenosine by an ecto-5’-nucleotidase expressed on macrophages, to activate the macrophage A2a adenosine receptor. Intraperitoneal injection of zymosan into Adora2a- or Panx1-deficient mice produced high, sustained levels of inflammatory mediators in the peritoneal lavage. These results indicated that AMP from apoptotic cells suppresses inflammation as a ‘calm down’ signal. DOI:http://dx.doi.org/10.7554/eLife.02172.001 Infections, toxins, and trauma can all injure tissue and cause the cells inside the tissue to die. When a cell dies, the membrane that surrounds it ruptures and its contents spill out, triggering inflammation of the surrounding tissues. This inflammation is part of the body’s efforts to begin the healing process but, if left uncontrolled, inflammation itself can cause further tissue damage. Diseased or damaged cells can also ‘choose’ to kill themselves to protect other healthy cells. This process, which is called apoptosis, also eliminates about 100,000 cells that are too old, or just no longer needed, from the human body every second. A cell undergoing apoptosis essentially dismantles itself, and the remains of the cell are packaged up, and cleared away by the white blood cells. Interestingly, this programed cell death releases many of the same molecules as other dying cells, but does so without triggering inflammation. The reason behind this lack of inflammation has not been clear. Now, Yamaguchi et al. have addressed this issue, and shown that cells undergoing apoptosis also release a chemical called adenosine monophosphate (AMP) that acts as a ‘calm down’ signal. The AMP is processed by white blood cells to a simpler chemical, which ‘switches on’ various genes in the white blood cells. This leads to the production of proteins that suppress the inflammation that would otherwise be triggered by other molecules released from the cells undergoing apoptosis. The findings of Yamaguchi et al. show how the community of cells in our body is kept in a healthy balance, and in the future, could improve our understanding and the treatment of inflammatory diseases. DOI:http://dx.doi.org/10.7554/eLife.02172.002
Collapse
Affiliation(s)
- Hiroshi Yamaguchi
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | |
Collapse
|
231
|
Brentnall M, Weir DB, Rongvaux A, Marcus AI, Boise LH. Procaspase-3 regulates fibronectin secretion and influences adhesion, migration and survival independently of catalytic function. J Cell Sci 2014; 127:2217-26. [PMID: 24610949 DOI: 10.1242/jcs.135137] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Caspase-3 is an effector caspase that is activated downstream of mitochondrial outer-membrane permeabilization (MOMP) during apoptosis. However, previous work has demonstrated that caspase-3-deficient mouse embryonic fibroblasts (MEFs) are resistant to mitochondrially mediated cell death and display a delay in the mitochondrial events of apoptosis, including Bax activation, MOMP and release of cytochrome c. Here, we show that caspase-3 regulates fibronectin secretion and impacts on cell morphology, adhesion and migration. Surprisingly, the catalytic activity of caspase-3 is not required for these non-apoptotic functions. Moreover, we found that caspase-3-deficient MEFs are not resistant to death by anoikis and that exogenous fibronectin protects wild-type MEFs from cell death induced by serum withdrawal. Taken together, our data indicate that procaspase-3 has a non-apoptotic function; it regulates the secretion of fibronectin and influences morphology, adhesion and migration. Furthermore, this novel procaspase-3 function might alter the apoptotic threshold of the cell.
Collapse
Affiliation(s)
- Matthew Brentnall
- Departments of Hematology and Medical Oncology and Cell Biology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David B Weir
- Cancer Biology Graduate Program, Emory University, Atlanta, GA 30322, USA
| | - Anthony Rongvaux
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Adam I Marcus
- Departments of Hematology and Medical Oncology and Cell Biology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Lawrence H Boise
- Departments of Hematology and Medical Oncology and Cell Biology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
232
|
Fan Y, Wang S, Hernandez J, Yenigun VB, Hertlein G, Fogarty CE, Lindblad JL, Bergmann A. Genetic models of apoptosis-induced proliferation decipher activation of JNK and identify a requirement of EGFR signaling for tissue regenerative responses in Drosophila. PLoS Genet 2014; 10:e1004131. [PMID: 24497843 PMCID: PMC3907308 DOI: 10.1371/journal.pgen.1004131] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 12/06/2013] [Indexed: 11/19/2022] Open
Abstract
Recent work in several model organisms has revealed that apoptotic cells are able to stimulate neighboring surviving cells to undergo additional proliferation, a phenomenon termed apoptosis-induced proliferation. This process depends critically on apoptotic caspases such as Dronc, the Caspase-9 ortholog in Drosophila, and may have important implications for tumorigenesis. While it is known that Dronc can induce the activity of Jun N-terminal kinase (JNK) for apoptosis-induced proliferation, the mechanistic details of this activation are largely unknown. It is also controversial if JNK activity occurs in dying or in surviving cells. Signaling molecules of the Wnt and BMP families have been implicated in apoptosis-induced proliferation, but it is unclear if they are the only ones. To address these questions, we have developed an efficient assay for screening and identification of genes that regulate or mediate apoptosis-induced proliferation. We have identified a subset of genes acting upstream of JNK activity including Rho1. We also demonstrate that JNK activation occurs both in apoptotic cells as well as in neighboring surviving cells. In a genetic screen, we identified signaling by the EGFR pathway as important for apoptosis-induced proliferation acting downstream of JNK signaling. These data underscore the importance of genetic screening and promise an improved understanding of the mechanisms of apoptosis-induced proliferation. Work in recent years has revealed that apoptotic caspases not only induce apoptosis, but also have non-apoptotic functions. One of these functions is apoptosis-induced proliferation, a relatively recently discovered phenomenon by which apoptotic cells induce proliferation of surviving neighboring cells. This phenomenon may have important implications for stem cell activity, tissue regeneration and tumorigenesis. Here, we describe the development of a genetic model of apoptosis-induced proliferation and the use of this model for convenient and unbiased genetic screening to identify genes involved in the process. We tested mutants of our RNAi transgenic lines targeting the core components of the apoptotic pathway and of JNK signaling, a known mediator of apoptosis-induced proliferation. These assays demonstrate the feasibility of the system for systematic genetic screening and identified several new genes upstream of JNK that are involved in apoptosis-induced proliferation. Finally, we tested the model in a pilot screen for chromosome arm 2L and identified spi, the EGF ligand in flies, as important for apoptosis-induced proliferation. We confirmed the involvement of EGF in a genuine apoptosis-induced regeneration system. These data underscore the importance of genetic screening and promise an improved understanding of the mechanisms of apoptosis-induced proliferation and regeneration.
Collapse
Affiliation(s)
- Yun Fan
- University of Massachusetts Medical School, Department of Cancer Biology, Worcester, Massachusetts, United States of America
- * E-mail: (YF); (AB)
| | - Shiuan Wang
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jacob Hernandez
- MD Anderson Cancer Center, Department of Biochemistry & Molecular Biology, Houston, Texas, United States of America
| | - Vildan Betul Yenigun
- MD Anderson Cancer Center, Department of Biochemistry & Molecular Biology, Houston, Texas, United States of America
| | - Gillian Hertlein
- Länderinstitut für Bienenkunde, Humboldt Universität zu Berlin, Hohen Neuendorf, Germany
| | - Caitlin E. Fogarty
- University of Massachusetts Medical School, Department of Cancer Biology, Worcester, Massachusetts, United States of America
| | - Jillian L. Lindblad
- University of Massachusetts Medical School, Department of Cancer Biology, Worcester, Massachusetts, United States of America
| | - Andreas Bergmann
- University of Massachusetts Medical School, Department of Cancer Biology, Worcester, Massachusetts, United States of America
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- MD Anderson Cancer Center, Department of Biochemistry & Molecular Biology, Houston, Texas, United States of America
- * E-mail: (YF); (AB)
| |
Collapse
|
233
|
Abstract
In many animals, regenerative processes can replace lost body parts. Organ and tissue regeneration consequently also hold great medical promise. The regulation of regenerative processes is achieved through concerted actions of multiple organizational levels of the organism, from diffusing molecules and cellular gene expression patterns up to tissue mechanics. Our intuition is usually not adapted well to this degree of complexity and the quantitative aspects of the regulation of regenerative processes remain poorly understood. One way out of this dilemma lies in the combination of experimentation and mathematical modeling within an iterative process of model development/refinement, model predictions for novel experimental conditions, quantitative experiments testing these predictions, and subsequent model refinement. This interdisciplinary approach has already provided key insights into smaller scale processes during embryonic development and a so-far limited number of more complex regeneration processes. This review discusses selected theoretical and interdisciplinary studies and is structured along the three phases of regeneration: (1) initiation of a regeneration response, (2) tissue patterning during regenerate growth, (3) arresting the regeneration response. Moreover, we highlight the opportunities provided by extensions of mathematical models from developmental processes toward the study of related regenerative processes.
Collapse
Affiliation(s)
- Osvaldo Chara
- Center for Information Services and High Performance Computing (ZIH), Technische Universität Dresden, Dresden, Germany
| | - Elly M Tanaka
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Lutz Brusch
- Center for Information Services and High Performance Computing (ZIH), Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
234
|
Abstract
Regeneration is a process by which organisms replace damaged or amputated organs to restore normal body parts. Regeneration of many tissues or organs requires proliferation of stem cells or stem cell-like blastema cells. This regenerative growth is often initiated by cell death pathways induced by damage. The executors of regenerative growth are a group of growth-promoting signaling pathways, including JAK/STAT, EGFR, Hippo/YAP, and Wnt/β-catenin. These pathways are also essential to developmental growth, but in regeneration, they are activated in distinct ways and often at higher strengths, under the regulation by certain stress-responsive signaling pathways, including JNK signaling. Growth suppressors are important in termination of regeneration to prevent unlimited growth and also contribute to the loss of regenerative capacity in nonregenerative organs. Here, we review cellular and molecular growth regulation mechanisms induced by organ damage in several models with different regenerative capacities.
Collapse
Affiliation(s)
- Gongping Sun
- Howard Hughes Medical Institute, Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, Piscataway, New Jersey, USA
| | - Kenneth D Irvine
- Howard Hughes Medical Institute, Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers The State University of New Jersey, Piscataway, New Jersey, USA.
| |
Collapse
|
235
|
|
236
|
Gauron C, Rampon C, Bouzaffour M, Ipendey E, Teillon J, Volovitch M, Vriz S. Sustained production of ROS triggers compensatory proliferation and is required for regeneration to proceed. Sci Rep 2013; 3:2084. [PMID: 23803955 PMCID: PMC3694286 DOI: 10.1038/srep02084] [Citation(s) in RCA: 219] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 06/10/2013] [Indexed: 12/23/2022] Open
Abstract
A major issue in regenerative medicine is the role of injury in promoting cell plasticity. Here we explore the function of reactive oxygen species (ROS) induced through lesions in adult zebrafish. We show that ROS production, following adult fin amputation, is tightly regulated in time and space for at least 24 hours, whereas ROS production remains transient (2 hours) in mere wound healing. In regenerative tissue, ROS signaling triggers two distinct parallel pathways: one pathway is responsible for apoptosis, and the other pathway is responsible for JNK activation. Both events are involved in the compensatory proliferation of stump epidermal cells and are necessary for the progression of regeneration. Both events impact the Wnt, SDF1 and IGF pathways, while apoptosis only impacts progenitor marker expression. These results implicate oxidative stress in regeneration and provide new insights into the differences between healing and regeneration.
Collapse
Affiliation(s)
- Carole Gauron
- Centre Interdisciplinaire de Recherche en Biologie-CIRB, CNRS UMR 7241/INSERM U1050/Collège de France, Paris, France
| | | | | | | | | | | | | |
Collapse
|
237
|
Palechor-Ceron N, Suprynowicz FA, Upadhyay G, Dakic A, Minas T, Simic V, Johnson M, Albanese C, Schlegel R, Liu X. Radiation induces diffusible feeder cell factor(s) that cooperate with ROCK inhibitor to conditionally reprogram and immortalize epithelial cells. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1862-1870. [PMID: 24096078 DOI: 10.1016/j.ajpath.2013.08.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/31/2013] [Accepted: 08/26/2013] [Indexed: 10/26/2022]
Abstract
Both feeder cells and Rho kinase inhibition are required for the conditional reprogramming and immortalization of human epithelial cells. In the present study, we demonstrated that the Rho kinase inhibitor Y-27632, significantly suppresses keratinocyte differentiation and extends life span in serum-containing medium but does not lead to immortalization in the absence of feeder cells. Using Transwell culture plates, we further demonstrated that physical contact between the feeder cells and keratinocytes is not required for inducing immortalization and, more importantly, that irradiation of the feeder cells is required for this induction. Consistent with these experiments, conditioned medium was shown to induce and maintain conditionally immortalized cells, which was accompanied by increased telomerase expression. The activity of conditioned medium directly correlated with radiation-induced apoptosis of the feeder cells. Thus, the induction of conditionally reprogrammed cells is mediated by a combination of Y-27632 and a diffusible factor (or factors) released by apoptotic feeder cells.
Collapse
Affiliation(s)
- Nancy Palechor-Ceron
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia
| | - Frank A Suprynowicz
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia
| | - Geeta Upadhyay
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia
| | - Aleksandra Dakic
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia
| | - Tsion Minas
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia
| | - Vera Simic
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia
| | - Michael Johnson
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Christopher Albanese
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia; Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Richard Schlegel
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia.
| | - Xuefeng Liu
- Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia; Center for Cellular Reprogramming, Georgetown University Medical Center, Washington, District of Columbia.
| |
Collapse
|
238
|
Pérez-Garijo A, Fuchs Y, Steller H. Apoptotic cells can induce non-autonomous apoptosis through the TNF pathway. eLife 2013; 2:e01004. [PMID: 24066226 PMCID: PMC3779319 DOI: 10.7554/elife.01004] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 08/06/2013] [Indexed: 12/14/2022] Open
Abstract
Apoptotic cells can produce signals to instruct cells in their local environment, including ones that stimulate engulfment and proliferation. We identified a novel mode of communication by which apoptotic cells induce additional apoptosis in the same tissue. Strong induction of apoptosis in one compartment of the Drosophila wing disc causes apoptosis of cells in the other compartment, indicating that dying cells can release long-range death factors. We identified Eiger, the Drosophila tumor necrosis factor (TNF) homolog, as the signal responsible for apoptosis-induced apoptosis (AiA). Eiger is produced in apoptotic cells and, through activation of the c-Jun N-terminal kinase (JNK) pathway, is able to propagate the initial apoptotic stimulus. We also show that during coordinated cell death of hair follicle cells in mice, TNF-α is expressed in apoptotic cells and is required for normal cell death. AiA provides a mechanism to explain cohort behavior of dying cells that is seen both in normal development and under pathological conditions. DOI:http://dx.doi.org/10.7554/eLife.01004.001 The tissues of developing organisms can be shaped by apoptosis, a form of regulated cell killing. Although this process can occur in individual cells, apoptotic signals may also dictate the ‘communal death’ of many cells simultaneously. This occurs frequently in animal development: in human fetuses, for example, cells in the hand are directed to die to remove webbing between the fingers. Apoptosis has been thought to resemble a form of silent suicide by cells, but more recent work suggests that apoptotic cells can also transmit signals. Now, Pérez-Garijo et al. find that these cells can stimulate other cells to die in both fruit flies and mice. In fruit flies, apoptosis is activated by proteins known as Grim, Hid and Reaper. To explore whether apoptotic cells could communicate with other cells, Pérez-Garijo et al. created ‘undead’ cells in which one of these proteins was turned on, but other downstream proteins (that are responsible for the cellular execution phase of apoptosis) had been turned off: these cells were undergoing apoptosis, but could not complete the process and die. Strikingly, undead cells in the posterior (back) region of the wing imaginal disc—the tissue in the larva that gives rise to the wing in the adult fruit fly—could trigger apoptosis in cells in the anterior (front) half. Pérez-Garijo et al. found that the JNK pathway activated apoptosis in anterior cells. In fruit flies, the Eiger protein turns on this pathway; when Eiger was absent from posterior cells in the wing imaginal disc, apoptosis in anterior cells ceased, indicating that Eiger might signal at long range. Eiger is related to a protein called TNF that has been implicated in cycles of destruction and renewal of hair follicles in mice. Pérez-Garijo et al. found that TNF is produced by apoptotic cells in hair follicles, and that blocking TNF inhibits the death of other cells in the same cohort: this suggests that a common mechanism could regulate the communal death of cells in flies and mammals. These studies therefore shed light on a conserved pathway in the modulation of tissue development. DOI:http://dx.doi.org/10.7554/eLife.01004.002
Collapse
Affiliation(s)
- Ainhoa Pérez-Garijo
- Strang Laboratory of Apoptosis and Cancer Biology , Howard Hughes Medical Institute, The Rockefeller University , New York , United States
| | | | | |
Collapse
|
239
|
Boland K, Flanagan L, Prehn JHM. Paracrine control of tissue regeneration and cell proliferation by Caspase-3. Cell Death Dis 2013; 4:e725. [PMID: 23846227 PMCID: PMC3730423 DOI: 10.1038/cddis.2013.250] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 06/07/2013] [Indexed: 02/06/2023]
Abstract
Executioner caspases such as Caspase-3 and Caspase-7 have long been recognised as the key proteases involved in cell demolition during apoptosis. Caspase activation also modulates signal transduction inside cells, through activation or inactivation of kinases, phosphatases and other signalling molecules. Interestingly, a series of recent studies have demonstrated that caspase activation may also influence signal transduction and gene expression changes in neighbouring cells that themselves did not activate caspases. This review describes the physiological relevance of paracrine Caspase-3 signalling for developmental processes, tissue homeostasis and tissue regeneration, and discusses the role of soluble factors and microparticles in mediating these paracrine activities. While non-cell autonomous control of tissue regeneration by Caspase-3 may represent an important process for maintaining tissue homeostasis, it may limit the efficiency of current cancer therapy by promoting cell proliferation in those cancer cells resistant to radio- or chemotherapy. We discuss recent evidence in support of such a role for Caspase-3, and discuss its therapeutic implication.
Collapse
Affiliation(s)
- K Boland
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | | | | |
Collapse
|
240
|
Gómez-Orte E, Sáenz-Narciso B, Moreno S, Cabello J. Multiple functions of the noncanonical Wnt pathway. Trends Genet 2013; 29:545-53. [PMID: 23846023 DOI: 10.1016/j.tig.2013.06.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/29/2013] [Accepted: 06/07/2013] [Indexed: 01/11/2023]
Abstract
Thirty years after the identification of WNTs, understanding of their signal transduction pathways continues to expand. Here, we review recent advances in characterizing the Wnt-dependent signaling pathways in Caenorhabditis elegans linking polar signals to rearrangements of the cytoskeleton in different developmental processes, such as proper mitotic spindle orientation, cell migration, and engulfment of apoptotic corpses. In addition to the well-described transcriptional outputs of the canonical and noncanonical Wnt pathways, new branches regulating nontranscriptional outputs that control RAC (Ras related GTPase) activity are also discussed. These findings suggest that Wnt signaling is a master regulator not only of development, but also of cell polarization.
Collapse
Affiliation(s)
- Eva Gómez-Orte
- Center for Biomedical Research of La Rioja (CIBIR), C/Piqueras 98, 26006 Logroño, Spain
| | | | | | | |
Collapse
|
241
|
Herrera SC, Martín R, Morata G. Tissue homeostasis in the wing disc of Drosophila melanogaster: immediate response to massive damage during development. PLoS Genet 2013; 9:e1003446. [PMID: 23633961 PMCID: PMC3636033 DOI: 10.1371/journal.pgen.1003446] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 02/27/2013] [Indexed: 11/18/2022] Open
Abstract
All organisms have developed mechanisms to respond to organ or tissue damage that may appear during development or during the adult life. This process of regeneration is a major long-standing problem in Developmental Biology. We are using the Drosophila melanogaster wing imaginal disc to study the response to major damage inflicted during development. Using the Gal4/UAS/Gal80TS conditional system, we have induced massive cell killing by forcing activity of the pro-apoptotic gene hid in two major regions of the disc as defined by Gal4 inserts in the genes rotund (rn) and spalt (sal). The procedure ensures that at the end of a 40–48 hrs of ablation period the great majority of the cells of the original Rn or Sal domains have been eliminated. The results indicate that the damage provokes an immediate response aimed to keep the integrity of the epithelium and to repair the region under ablation. This includes an increase in cell proliferation to compensate for the cell loss and the replacement of the dead cells by others from outside of the damaged area. The response is almost contemporaneous with the damage, so that at the end of the ablation period the targeted region is already reconstructed. We find that the proliferative response is largely systemic, as the number of cells in division increases all over the disc. Furthermore, our results indicate that the Dpp and Wg pathways are not specifically involved in the regenerative response, but that activity of the JNK pathway is necessary both inside and outside the ablated domain for its reconstruction. The study of how organs or tissues regenerate after damage is a classic topic in Developmental Biology. We are studying this process in the developing wing imaginal disc of Drosophila melanogaster, using genetic methods to inflict massive damage in the region destined to form the wing blade. We find that the lesion provokes a very strong and rapid reaction in the remaining disc aimed to reconstruct the lost tissue, both in size and in shape. The response includes an increase of cell proliferation to compensate for the loss of cells and the immigration of cells from neighbouring areas to replace the dead ones. The immigrant cells change their original identity and acquire that of the cells they are replacing. We propose that these experiments reveal the existence of a powerful homeostatic mechanism that is able to cure massive injuries that may appear during development.
Collapse
Affiliation(s)
- Salvador C. Herrera
- Centro de Biología Molecular CSIC–UAM, Universidad Autónoma de Madrid, Madrid, Spain
| | - Raquel Martín
- Centro de Biología Molecular CSIC–UAM, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ginés Morata
- Centro de Biología Molecular CSIC–UAM, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
242
|
Homeostatic epithelial renewal in the gut is required for dampening a fatal systemic wound response in Drosophila. Cell Rep 2013; 3:919-30. [PMID: 23523355 DOI: 10.1016/j.celrep.2013.02.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 11/08/2012] [Accepted: 02/19/2013] [Indexed: 11/21/2022] Open
Abstract
Effective defense responses involve the entire organism. To maintain body homeostasis after tissue damage, a systemic wound response is induced in which the response of each tissue is tightly orchestrated to avoid incomplete recovery or an excessive, damaging response. Here, we provide evidence that in the systemic response to wounding, an apoptotic caspase pathway is activated downstream of reactive oxygen species in the midgut enterocytes (ECs), cells distant from the wound site, in Drosophila. We show that a caspase-pathway mutant has defects in homeostatic gut cell renewal and that inhibiting caspase activity in fly ECs results in the production of systemic lethal factors after wounding. Our results indicate that wounding remotely controls caspase activity in ECs, which activates the tissue stem cell regeneration pathway in the gut to dampen the dangerous systemic wound reaction.
Collapse
|
243
|
Galliot B. Injury-induced asymmetric cell death as a driving force for head regeneration in Hydra. Dev Genes Evol 2013; 223:39-52. [PMID: 22833103 DOI: 10.1007/s00427-012-0411-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 06/04/2012] [Indexed: 12/25/2022]
Abstract
The freshwater Hydra polyp provides a unique model system to decipher the mechanisms underlying adult regeneration. Indeed, a single cut initiates two distinct regenerative processes, foot regeneration on one side and head regeneration on the other side, the latter relying on the rapid formation of a local head organizer. Two aspects are discussed here: the asymmetric cellular remodeling induced by mid-gastric bisection and the signaling events that trigger head organizer formation. In head-regenerating tips (but not in foot ones), a wave of cell death takes place immediately, leading the apoptotic cells to transiently release Wnt3 and activate the β-catenin pathway in the neighboring cycling cells to push them through mitosis. This process, which mimics the apoptosis-induced compensatory proliferation process deciphered in Drosophila larvae regenerating their discs, likely corresponds to an evolutionarily conserved mechanism, also at work in Xenopus tadpoles regenerating their tail or mice regenerating their skin or liver. How is this process generated in Hydra? Several studies pointed to the necessary activation of the extracellular signal-regulated kinase (ERK) 1-2 and mitogen-activated protein kinase (MAPK) pathways during early head regeneration. Indeed inhibition of ERK 1-2 or knockdown of RSK, cAMP response element-binding protein (CREB), and CREB-binding protein (CBP) prevent injury-induced apoptosis and head regeneration. The current scenario involves an asymmetric activation of the MAPK/CREB pathway to trigger injury-induced apoptosis in the interstitial cells and in the epithelial cells a CREB/CBP-dependent transcriptional activation of early genes essential for head-organizing activity as wnt3, HyBra1, and prdl-a. The question now is how bisection in the rather uniform central region of the polyp can generate this immediately asymmetric signaling.
Collapse
Affiliation(s)
- Brigitte Galliot
- Department of Genetics and Evolution, University of Geneva, 30 quai Ernest Ansermet, CH-1211 Geneva-04, Switzerland.
| |
Collapse
|
244
|
Mao P, Smith L, Xie W, Wang M. Dying endothelial cells stimulate proliferation of malignant glioma cells via a caspase 3-mediated pathway. Oncol Lett 2013; 5:1615-1620. [PMID: 23760767 PMCID: PMC3678715 DOI: 10.3892/ol.2013.1223] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 02/07/2013] [Indexed: 01/22/2023] Open
Abstract
Emerging evidence has indicated that apoptotic cells have a compensatory effect on the proliferation of neighboring cells. However, the potential role of dying vascular endothelial cells (ECs) in glioma tumor proliferation remains unclear. In the present study, three glioma cell lines were cocultured with dying ECs under various conditions to evaluate the effect of dying ECs on tumor proliferation using alamarBlue and trypan blue assays to assess cell proliferation and viability, respectively. The results suggested that dying ECs had a marked ability to facilitate glioma cell growth via a caspase 3-mediated pathway. Furthermore, calcium-independent phospholipase A2 (iPLA2), a downstream gene regulated by caspase 3, is highly involved in this process. Prostaglandin E2 (PGE2) was the final effector of the caspase 3-iPLA2 signaling pathway in glioma cell proliferation. Knockdown of caspase 3 or iPLA2 using shRNA negated the growth stimulating effect of dying ECs. By contrast, the overexpression of iPLA2 in ECs via the pLEX lentiviral vector system or addition of PGE2 into culture medium had a growth promoting effect on glioma cells. Overall, the present data revealed a paracrine signal released from dying ECs which promotes the proliferation of surrounding glioma cells, demonstrating the importance of blocking compensatory proliferation during tumor therapy. Additionally, targeting caspase 3-mediated pathways combined with current therapeutic strategies may be a promising approach for improving the dismal prognosis associated with these malignant tumors.
Collapse
Affiliation(s)
- Ping Mao
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China ; ; Department of Neurological Surgery, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
245
|
Abstract
Programmed cell death is an important process during development that serves to remove superfluous cells and tissues, such as larval organs during metamorphosis, supernumerary cells during nervous system development, muscle patterning and cardiac morphogenesis. Different kinds of cell death have been observed and were originally classified based on distinct morphological features: (1) type I programmed cell death (PCD) or apoptosis is recognized by cell rounding, DNA fragmentation, externalization of phosphatidyl serine, caspase activation and the absence of inflammatory reaction, (2) type II PCD or autophagy is characterized by the presence of large vacuoles and the fact that cells can recover until very late in the process and (3) necrosis is associated with an uncontrolled release of the intracellular content after cell swelling and rupture of the membrane, which commonly induces an inflammatory response. In this review, we will focus exclusively on developmental cell death by apoptosis and its role in tissue remodeling.
Collapse
|
246
|
Beane WS, Morokuma J, Lemire JM, Levin M. Bioelectric signaling regulates head and organ size during planarian regeneration. Development 2013; 140:313-22. [PMID: 23250205 DOI: 10.1242/dev.086900] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A main goal of regenerative medicine is to replace lost or damaged tissues and organs with functional parts of the correct size and shape. But the proliferation of new cells is not sufficient; we will also need to understand how the scale and ultimate form of newly produced tissues are determined. Using the planarian model system, we report that membrane voltage-dependent bioelectric signaling determines both head size and organ scaling during regeneration. RNA interference of the H(+),K(+)-ATPase ion pump results in membrane hyperpolarization, which has no effect on the amount of new tissue (blastema) that is regenerated yet produces regenerates with tiny 'shrunken' heads and proportionally oversized pharynges. Our data show that this disproportionality results from a lack of the apoptosis required to adjust head and organ size and placement, highlighting apoptotic remodeling as the link between bioelectric signaling and the establishment of organ size during regeneration.
Collapse
Affiliation(s)
- Wendy Scott Beane
- Biology Department and Tufts Center for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155, USA
| | | | | | | |
Collapse
|
247
|
Chaurio R, Janko C, Schorn C, Maueröder C, Bilyy R, Gaipl U, Schett G, Berens C, Frey B, Munoz LE. UVB-irradiated apoptotic cells induce accelerated growth of co-implanted viable tumor cells in immune competent mice. Autoimmunity 2013. [DOI: 10.3109/08916934.2012.754433] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
248
|
|
249
|
Mercer SE, Cheng CH, Atkinson DL, Krcmery J, Guzman CE, Kent DT, Zukor K, Marx KA, Odelberg SJ, Simon HG. Multi-tissue microarray analysis identifies a molecular signature of regeneration. PLoS One 2012; 7:e52375. [PMID: 23300656 PMCID: PMC3530543 DOI: 10.1371/journal.pone.0052375] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 11/14/2012] [Indexed: 02/06/2023] Open
Abstract
The inability to functionally repair tissues that are lost as a consequence of disease or injury remains a significant challenge for regenerative medicine. The molecular and cellular processes involved in complete restoration of tissue architecture and function are expected to be complex and remain largely unknown. Unlike humans, certain salamanders can completely regenerate injured tissues and lost appendages without scar formation. A parsimonious hypothesis would predict that all of these regenerative activities are regulated, at least in part, by a common set of genes. To test this hypothesis and identify genes that might control conserved regenerative processes, we performed a comprehensive microarray analysis of the early regenerative response in five regeneration-competent tissues from the newt Notophthalmus viridescens. Consistent with this hypothesis, we established a molecular signature for regeneration that consists of common genes or gene family members that exhibit dynamic differential regulation during regeneration in multiple tissue types. These genes include members of the matrix metalloproteinase family and its regulators, extracellular matrix components, genes involved in controlling cytoskeleton dynamics, and a variety of immune response factors. Gene Ontology term enrichment analysis validated and supported their functional activities in conserved regenerative processes. Surprisingly, dendrogram clustering and RadViz classification also revealed that each regenerative tissue had its own unique temporal expression profile, pointing to an inherent tissue-specific regenerative gene program. These new findings demand a reconsideration of how we conceptualize regenerative processes and how we devise new strategies for regenerative medicine.
Collapse
Affiliation(s)
- Sarah E. Mercer
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine and Children’s Memorial Research Center, Chicago, Illinois, United States of America
| | - Chia-Ho Cheng
- Department of Chemistry, University of Massachusetts-Lowell, Lowell, Massachusetts, United States of America
| | - Donald L. Atkinson
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - Jennifer Krcmery
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine and Children’s Memorial Research Center, Chicago, Illinois, United States of America
| | - Claudia E. Guzman
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine and Children’s Memorial Research Center, Chicago, Illinois, United States of America
| | - David T. Kent
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - Katherine Zukor
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - Kenneth A. Marx
- Department of Chemistry, University of Massachusetts-Lowell, Lowell, Massachusetts, United States of America
| | - Shannon J. Odelberg
- Department of Internal Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - Hans-Georg Simon
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine and Children’s Memorial Research Center, Chicago, Illinois, United States of America
| |
Collapse
|
250
|
In vivo cell and tissue dynamics underlying zebrafish fin fold regeneration. PLoS One 2012; 7:e51766. [PMID: 23284763 PMCID: PMC3527495 DOI: 10.1371/journal.pone.0051766] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/07/2012] [Indexed: 01/28/2023] Open
Abstract
Background Zebrafish (Danio rerio) has a remarkable capacity to regenerate many organs and tissues. During larval stages the fin fold allows the possibility of performing long time-lapse imaging making this system very appealing to study the relationships between tissue movements, cell migration and proliferation necessary for the regeneration process. Results Through the combined use of transgenic fluorescently-labeled animals and confocal microscopy imaging, we characterized in vivo the complete fin fold regeneration process. We show, for the first time, that there is an increase in the global rate of epidermal growth as a response to tissue loss. Also enhanced significantly is cell proliferation, which upon amputation happens in a broad area concerning the amputation level and not in a blastema-restricted way. This reveals a striking difference with regard to the adult fin regeneration system. Finally, an accumulation of migratory, shape-changing fibroblasts occurs proximally to the wound area, resembling a blastemal-like structure, which may act as a signaling center for the regeneration process to proceed. Conclusions These findings provide a novel in vivo description of fundamental mechanisms occurring during the fin fold regeneration process, thereby contributing to a better knowledge of this regenerative system and to reveal variations in the epimorphic regeneration field.
Collapse
|