1
|
Adell T, Cebrià F, Abril JF, Araújo SJ, Corominas M, Morey M, Serras F, González-Estévez C. Cell death in regeneration and cell turnover: Lessons from planarians and Drosophila. Semin Cell Dev Biol 2025; 169:103605. [PMID: 40139139 DOI: 10.1016/j.semcdb.2025.103605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/27/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
Programmed cell death plays a crucial role during tissue turnover in all animal species, and it is also essential during regeneration, serving as a key signalling mechanism to promote tissue repair and regrowth. In freshwater planarians, remarkable regenerative abilities are supported by neoblasts, a population of adult stem cells, which enable high somatic cell turnover. Cell death in planarians occurs continuously during regeneration and adult homeostasis, underscoring its critical role in tissue remodeling and repair. However, the exact mechanisms regulating cell death in these organisms remain elusive. In contrast, Drosophila melanogaster serves as a powerful model for studying programmed cell death in development, metamorphosis, and adult tissue maintenance, leveraging advanced genetic tools and visualization techniques. In Drosophila, cell death sculpts tissues, eliminates larval structures during metamorphosis, and supports homeostasis in adulthood. Despite limited regenerative capacity compared to planarians, Drosophila provides unique insights into cell death's regulatory mechanisms. Comparative analysis of these two systems highlights both conserved and divergent roles of programmed cell death in tissue renewal and regeneration. This review synthesizes the latest knowledge of programmed cell death in planarians and Drosophila, aiming to illuminate shared principles and system-specific adaptations, with relevance to tissue repair across biological systems.
Collapse
Affiliation(s)
- Teresa Adell
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain.
| | - Francesc Cebrià
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Josep F Abril
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Sofia J Araújo
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Montserrat Corominas
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Marta Morey
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Florenci Serras
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain
| | - Cristina González-Estévez
- Department of Genetics, Microbiology and Statistics, School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Av. Diagonal 643, Edifici Prevosti 1st floor, Barcelona 08028, Spain.
| |
Collapse
|
2
|
Cho CJ, Brown JW, Mills JC. Origins of cancer: ain't it just mature cells misbehaving? EMBO J 2024; 43:2530-2551. [PMID: 38773319 PMCID: PMC11217308 DOI: 10.1038/s44318-024-00099-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 05/23/2024] Open
Abstract
A pervasive view is that undifferentiated stem cells are alone responsible for generating all other cells and are the origins of cancer. However, emerging evidence demonstrates fully differentiated cells are plastic, can be coaxed to proliferate, and also play essential roles in tissue maintenance, regeneration, and tumorigenesis. Here, we review the mechanisms governing how differentiated cells become cancer cells. First, we examine the unique characteristics of differentiated cell division, focusing on why differentiated cells are more susceptible than stem cells to accumulating mutations. Next, we investigate why the evolution of multicellularity in animals likely required plastic differentiated cells that maintain the capacity to return to the cell cycle and required the tumor suppressor p53. Finally, we examine an example of an evolutionarily conserved program for the plasticity of differentiated cells, paligenosis, which helps explain the origins of cancers that arise in adults. Altogether, we highlight new perspectives for understanding the development of cancer and new strategies for preventing carcinogenic cellular transformations from occurring.
Collapse
Affiliation(s)
- Charles J Cho
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey W Brown
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
McCusker P, Clarke NG, Gardiner E, Armstrong R, McCammick EM, McVeigh P, Robb E, Wells D, Nowak-Roddy M, Albaqami A, Mousley A, Coulter JA, Harrington J, Marks NJ, Maule AG. Neoblast-like stem cells of Fasciola hepatica. PLoS Pathog 2024; 20:e1011903. [PMID: 38805551 PMCID: PMC11161113 DOI: 10.1371/journal.ppat.1011903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/07/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024] Open
Abstract
The common liver fluke (Fasciola hepatica) causes the disease fasciolosis, which results in considerable losses within the global agri-food industry. There is a shortfall in the drugs that are effective against both the adult and juvenile life stages within the mammalian host, such that new drug targets are needed. Over the last decade the stem cells of parasitic flatworms have emerged as reservoirs of putative novel targets due to their role in development and homeostasis, including at host-parasite interfaces. Here, we investigate and characterise the proliferating cells that underpin development in F. hepatica. We provide evidence that these cells are capable of self-renewal, differentiation, and are sensitive to ionising radiation- all attributes of neoblasts in other flatworms. Changes in cell proliferation were also noted during the early stages of in vitro juvenile growth/development (around four to seven days post excystment), which coincided with a marked reduction in the nuclear area of proliferating cells. Furthermore, we generated transcriptomes from worms following irradiation-based ablation of neoblasts, identifying 124 significantly downregulated transcripts, including known stem cell markers such as fgfrA and plk1. Sixty-eight of these had homologues associated with neoblast-like cells in Schistosoma mansoni. Finally, RNA interference mediated knockdown of histone h2b (a marker of proliferating cells), ablated neoblast-like cells and impaired worm development in vitro. In summary, this work demonstrates that the proliferating cells of F. hepatica are equivalent to neoblasts of other flatworm species and demonstrate that they may serve as attractive targets for novel anthelmintics.
Collapse
Affiliation(s)
- Paul McCusker
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Nathan G. Clarke
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Erica Gardiner
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Rebecca Armstrong
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Erin M. McCammick
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Paul McVeigh
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Emily Robb
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Duncan Wells
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Madelyn Nowak-Roddy
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Abdullah Albaqami
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Angela Mousley
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | | | - John Harrington
- Boehringer Ingelheim Animal Health, Duluth, Georgia, United States of America
| | - Nikki J. Marks
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Aaron G. Maule
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
4
|
Ko JM, Reginato W, Wolff A, Lobo D. Mechanistic regulation of planarian shape during growth and degrowth. Development 2024; 151:dev202353. [PMID: 38619319 PMCID: PMC11128284 DOI: 10.1242/dev.202353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Adult planarians can grow when fed and degrow (shrink) when starved while maintaining their whole-body shape. It is unknown how the morphogens patterning the planarian axes are coordinated during feeding and starvation or how they modulate the necessary differential tissue growth or degrowth. Here, we investigate the dynamics of planarian shape together with a theoretical study of the mechanisms regulating whole-body proportions and shape. We found that the planarian body proportions scale isometrically following similar linear rates during growth and degrowth, but that fed worms are significantly wider than starved worms. By combining a descriptive model of planarian shape and size with a mechanistic model of anterior-posterior and medio-lateral signaling calibrated with a novel parameter optimization methodology, we theoretically demonstrate that the feedback loop between these positional information signals and the shape they control can regulate the planarian whole-body shape during growth. Furthermore, the computational model produced the correct shape and size dynamics during degrowth as a result of a predicted increase in apoptosis rate and pole signal during starvation. These results offer mechanistic insights into the dynamic regulation of whole-body morphologies.
Collapse
Affiliation(s)
- Jason M. Ko
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Waverly Reginato
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Andrew Wolff
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Daniel Lobo
- Department of Biological Sciences, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
- Greenebaum Comprehensive Cancer Center and Center for Stem Cell Biology & Regenerative Medicine, University of Maryland, School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
5
|
Kang J, Ai Q, Zhao A, Wang H, Zhang X, Liu Y, Zhang L, Liu Y. Neurotoxicological mechanisms of carbon quantum dots in a new animal model Dugesia japonica. ECOTOXICOLOGY (LONDON, ENGLAND) 2023; 32:711-719. [PMID: 37386302 DOI: 10.1007/s10646-023-02671-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 07/01/2023]
Abstract
As luminescent nanomaterials, the carbon quantum dots (CQDs) research focused on emerging applications since their discovery. However, their toxicological effects on the natural environment are still unclear. The freshwater planarian Dugesia japonica is distributed extensively in aquatic ecosystems and can regenerate a new brain in 5 days after amputation. Therefore it can be used as a new model organism in the field of neuroregeneration toxicology. In our study, D. japonica was cut and incubated in medium treated with CQDs. The results showed that the injured planarian lost the neuronal ability of brain regeneration after treatment with CQDs. Its Hh signalling system was interfered with at Day 5, and all cultured pieces died on or before Day 10 due to head lysis. Our work reveals that CQDs might affect the nerve regeneration of freshwater planarians via the Hh signalling pathway. The results of this study improve our understanding of CQD neuronal development toxicology and can aid in the development of warning systems for aquatic ecosystem damage.
Collapse
Affiliation(s)
- Jing Kang
- College of Life Science, Xinxiang Medical University, Xinxiang, China.
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China.
| | - Qing Ai
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Ang Zhao
- College of Life Science, Xinxiang Medical University, Xinxiang, China
| | - Haijiao Wang
- College of Life Science, Xinxiang Medical University, Xinxiang, China
| | - Xiangpeng Zhang
- Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, China
| | - Yanli Liu
- College of Life Science, Xinxiang Medical University, Xinxiang, China
| | | | - Yuqing Liu
- College of Life Science, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
6
|
Halim Harrath A, Dahmash W, Alrezaki A, Mansour L, Alwasel S. Using autophagy, apoptosis, cytoskeleton, and epigenetics markers to investigate the origin of infertility in ex-fissiparous freshwater planarian individuals (nomen nudum species) with hyperplasic ovaries. J Invertebr Pathol 2023:107935. [PMID: 37209811 DOI: 10.1016/j.jip.2023.107935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 05/22/2023]
Abstract
The origin of the sterility observed in ex-fissiparous freshwater planarians with hyperplasic ovaries has yet to be explained. To improve our understanding of this enigmatic phenomenon, immunofluorescence staining and confocal microscopy examination were used the assess autophagy, apoptosis, cytoskeleton, and epigenetics markers in the hyperplasic ovaries of ex-fissiparous individuals and the normal ovaries of sexual individuals. Immunofluorescence positivity for the autophagic marker microtubule-associated protein1 light chain 3 (LC3) was significantly lower in the hyperplasic ovary than in the normal ovary. Compared with the normal ovary, the hyperplasic ovary exhibited significantly higher immunofluorescence positivity for the apoptotic marker caspase 3, suggesting that autophagy and apoptosis are closely associated in this pathogenicity. Furthermore, the level of global DNA (cytosine-5)-methyltransferase 3A (DNMT3) protein expression was significantly higher in the normal ovary than in the hyperplasic ovary, suggesting that DNA methylation is involved in the infertility phenomenon. The cytoskeleton marker actin also exhibited relatively higher immunofluorescence intensity in the normal ovary than in the hyperplasic ovary, consistent with previous findings on the role of cytoskeleton architecture in oocyte maturation. These results help improve our understanding of the causes of infertility in ex-fissiparous planarians with hyperplasic ovaries and provide new insights that will facilitate future studies on this mysterious pathogenicity.
Collapse
Affiliation(s)
- Abdel Halim Harrath
- King Saud University, Department of Zoology, College of Science, Riyadh, Saudi Arabia.
| | - Waleed Dahmash
- King Saud University, Department of Zoology, College of Science, Riyadh, Saudi Arabia
| | - Abdelkarem Alrezaki
- King Saud University, Department of Zoology, College of Science, Riyadh, Saudi Arabia
| | - Lamjed Mansour
- King Saud University, Department of Zoology, College of Science, Riyadh, Saudi Arabia
| | - Saleh Alwasel
- King Saud University, Department of Zoology, College of Science, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Ye K, Liu X, Li D, Gao L, Zheng K, Qu J, Xing N, Yang F, Liu B, Li A, Pang Q. Extracellular matrix-regulator MMPA is required for the orderly proliferation of neoblasts and differentiation of ectodermal progenitor cells in the planarian Dugesia japonica. Biochem Biophys Res Commun 2023; 659:1-9. [PMID: 37030019 DOI: 10.1016/j.bbrc.2023.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023]
Abstract
Matrix metalloproteinases (MMPs) are members of a family of zinc-dependent metallopeptidase proteins that are widely found in plants, animals, and microorganisms. As the regulators of the extracellular matrix and basement membrane, MMPs play an important role in embryogenesis, development, innate immunity, and regeneration. However, the function of MMP family in planarian, a model for regeneration research, is still ambiguous. Here, we cloned 5 MMPs genes from Dugesia japonica and found that DjMMPA was associated with the process of regeneration, neoblasts cell maintenance confusion and destruction. Loss of DjMMPA led to homeostasis confusion and eventually death, owing to neoblasts proliferation disorder. Additionally, DjMMPA RNAi-treated animals had impaired regeneration after amputation. Furthermore, knockdown of DjMMPA had noticeable defects in cell differentiation of ectoderm, especially in eyes and neural progenitor cells, possibly by inhibiting Wnt signaling. Our results suggest that extracellular matrix-regulator MMPA is required for the orderly proliferation of neoblasts and differentiation of ectodermal progenitor cells in the planarian, which provide valuable information for further explorations into the molecular mechanism of MMPS, stem cells, and regeneration.
Collapse
|
8
|
Hojjatipour T, Sohani M, Maali A, Rostami S, Azad M. Aberrant DNA Methylation Status and mRNA Expression Level of SMG1 Gene in Chronic Myeloid Leukemia: A Case-Control Study. CELL JOURNAL 2022; 24. [PMID: 36527348 PMCID: PMC9790066 DOI: 10.22074/cellj.2022.8526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
UNLABELLED OObjective: Chronic myeloid leukemia (CML) is a myeloproliferative malignancy with different stages. Aberrant epigenetic modifications, such as DNA methylation, have been introduced as a signature for diverse cancers which also plays a crucial role in CML pathogenesis and development. Suppressor with morphogenetic effect on genitalia (SMG1) gene recently has been brought to the spotlight as a potent tumor suppressor gene that can be suppressed by tumors for further progress. The present study aims to investigate SMG1 status in CML patients. MATERIALS AND METHODS In this case-control study, peripheral blood from 30 patients with different phases of CML [new case (N)=10, complete molecular remission (CMR)=10, blastic phase (BP)=10] and 10 healthy subjects were collected. Methylation status and expression level of SMG1 gene promoter was assessed by methylation-specific polymerase chain reaction (MSP) and quantitative reverse-transcription PCR, respectively. RESULTS MSP results of SMG1 gene promotor in the new case group were methylated (60% methylated, 30% hemimethylated and 10% unmethylated). All CMR and control group patients were unmethylated in the SMG1 gene promoter. In the BP group, methylated SMG1 promoter was seen (50% of patients had a methylated status and 50% had hemimethylated status). In comparison with the healthy subjects, expression level of SMG1 in the new case group was decreased (P<0.01); in the CMR group and BP-CML groups, it was increased (P<0.05). No significant correlation between patients' hematological features and SMG1 methylation was seen. CONCLUSION Our results demonstrated that aberrant methylation of SMG1 occurred in CML patients and it had a significant association with SMG1 expression. SMG1 gene promoter showed diverse methylated status and subsequent expression levels in different phases of CML. These findings suggested possible participation of SMG1 suppression in the CML pathogenesis.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Department of Hematology and Blood Transfusion, Students Research Center, School of Allied Medicine, Tehran University of Medical
Sciences, Tehran, Iran
| | - Mahsa Sohani
- Department of Hematology and Blood Transfusion, Students Research Center, School of Allied Medicine, Tehran University of Medical
Sciences, Tehran, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, School of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Shahrbano Rostami
- Hematologic Malignancies Research Center, Tehran University of Medical Sciences, Tehran, Iran,P.O.Box: 3419915315Hematologic Malignancies Research CenterTehran University of Medical SciencesTehranIranP.O.Box: 1416634793Department of Medical Laboratory SciencesSchool of ParamedicineQazvin University of Medical SciencesQazvinIran
Emails:,
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Qazvin University of Medical Sciences, Qazvin, Iran,P.O.Box: 3419915315Hematologic Malignancies Research CenterTehran University of Medical SciencesTehranIranP.O.Box: 1416634793Department of Medical Laboratory SciencesSchool of ParamedicineQazvin University of Medical SciencesQazvinIran
Emails:,
| |
Collapse
|
9
|
PI(18:1/18:1) is a SCD1-derived lipokine that limits stress signaling. Nat Commun 2022; 13:2982. [PMID: 35624087 PMCID: PMC9142606 DOI: 10.1038/s41467-022-30374-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/27/2022] [Indexed: 02/07/2023] Open
Abstract
Cytotoxic stress activates stress-activated kinases, initiates adaptive mechanisms, including the unfolded protein response (UPR) and autophagy, and induces programmed cell death. Fatty acid unsaturation, controlled by stearoyl-CoA desaturase (SCD)1, prevents cytotoxic stress but the mechanisms are diffuse. Here, we show that 1,2-dioleoyl-sn-glycero-3-phospho-(1’-myo-inositol) [PI(18:1/18:1)] is a SCD1-derived signaling lipid, which inhibits p38 mitogen-activated protein kinase activation, counteracts UPR, endoplasmic reticulum-associated protein degradation, and apoptosis, regulates autophagy, and maintains cell morphology and proliferation. SCD1 expression and the cellular PI(18:1/18:1) proportion decrease during the onset of cell death, thereby repressing protein phosphatase 2 A and enhancing stress signaling. This counter-regulation applies to mechanistically diverse death-inducing conditions and is found in multiple human and mouse cell lines and tissues of Scd1-defective mice. PI(18:1/18:1) ratios reflect stress tolerance in tumorigenesis, chemoresistance, infection, high-fat diet, and immune aging. Together, PI(18:1/18:1) is a lipokine that links fatty acid unsaturation with stress responses, and its depletion evokes stress signaling. Fatty acid unsaturation by stearoyl-CoA desaturase 1 (SCD1) protects against cellular stress through unclear mechanisms. Here the authors show 1,2-dioleoyl-sn-glycero-3-phospho-(1’-myo-inositol) is an SCD1-derived signaling lipid that regulates stress-adaption, protects against cell death and promotes proliferation.
Collapse
|
10
|
Martinez P, Ballarin L, Ereskovsky AV, Gazave E, Hobmayer B, Manni L, Rottinger E, Sprecher SG, Tiozzo S, Varela-Coelho A, Rinkevich B. Articulating the "stem cell niche" paradigm through the lens of non-model aquatic invertebrates. BMC Biol 2022; 20:23. [PMID: 35057814 PMCID: PMC8781081 DOI: 10.1186/s12915-022-01230-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Stem cells (SCs) in vertebrates typically reside in "stem cell niches" (SCNs), morphologically restricted tissue microenvironments that are important for SC survival and proliferation. SCNs are broadly defined by properties including physical location, but in contrast to vertebrates and other "model" organisms, aquatic invertebrate SCs do not have clearly documented niche outlines or properties. Life strategies such as regeneration or asexual reproduction may have conditioned the niche architectural variability in aquatic or marine animal groups. By both establishing the invertebrates SCNs as independent types, yet allowing inclusiveness among them, the comparative analysis will allow the future functional characterization of SCNs.
Collapse
Affiliation(s)
- P Martinez
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
- Institut Català de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| | - L Ballarin
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35100, Padova, Italy
| | - A V Ereskovsky
- Aix Marseille University, Avignon Université, CNRS, IRD, IMBE, Marseille, France
- St. Petersburg State University, Biological Faculty, Universitetskaya emb. 7/9, St. Petersburg, 199034, Russia
- N. K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova Street 26, Moscow, 119334, Russia
| | - E Gazave
- Université de Paris, CNRS, Institut Jacques Monod, F-75006, Paris, France
| | - B Hobmayer
- Department of Zoology and Center of Molecular Biosciences, University of Innsbruck, Technikerstr. 25, 6020, Innsbruck, Austria
| | - L Manni
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35100, Padova, Italy
| | - E Rottinger
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
- Université Côte d'Azur, Federative Research Institute - Marine Resources (IFR MARRES), Nice, France
| | - S G Sprecher
- Department of Biology, University of Fribourg, Chemin du Musee 10, 1700, Fribourg, Switzerland
| | - S Tiozzo
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), Paris, France
| | - A Varela-Coelho
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Av. da República, 2780-157, Oeiras, Portugal
| | - B Rinkevich
- Israel Oceanography and Limnological Research, National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, 31080, Haifa, Israel.
| |
Collapse
|
11
|
Kang J, Chen J, Dong Z, Chen G, Liu D. The negative effect of the PI3K inhibitor 3-methyladenine on planarian regeneration via the autophagy signalling pathway. ECOTOXICOLOGY (LONDON, ENGLAND) 2021; 30:1941-1948. [PMID: 34403000 DOI: 10.1007/s10646-021-02439-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 06/13/2023]
Abstract
As an important PI3K (VPS34) inhibitor, 3-methyladenine (3-MA) can block the formation of autophagic vesicles in animals. Most toxicological studies using 3-MA have shown that 3-MA leads to serious disorders via autophagy suppression in mammals. However, no toxicological research on 3-MA has been performed on individuals undergoing regeneration. The freshwater planarian has powerful regenerative capability, and it can regenerate a new brain in 5 days and undergo complete adult individual remodelling in approximately 14 days. Moreover, it is also an excellent model organism for studies on environmental toxicology due to its high chemical sensitivity and extensive distribution. Here, Dugesia japonica planarians were treated with 3-MA, and the results showed that autophagy was inhibited and Djvps34 expression levels were down-regulated. After exposure to 10 mM 3-MA for 18 h, all the controls showed normal phenotypes, while one-half of the planarians treated with 3-MA showed morphological defects. In most cases, an ulcer appeared in the middle of the body, and a normal phenotype was restored 7 days following 3-MA exposure. During regeneration, disproportionate blastemas with tissue regression were observed. Furthermore, 3-MA treatment suppressed stem cell proliferation in intact and regenerating worms. These findings demonstrate that autophagy is indispensable for tissue homeostasis and regeneration in planarians and that 3-MA treatment is detrimental to planarian regeneration via its effect on the autophagy pathway.
Collapse
Affiliation(s)
- Jing Kang
- College of Life Science, Henan Normal University, Xinxiang, China
- College of Life Science, Xingxiang Medical University, Xinxiang, China
| | - Jinzi Chen
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Zimei Dong
- College of Life Science, Henan Normal University, Xinxiang, China.
| | - Guangwen Chen
- College of Life Science, Henan Normal University, Xinxiang, China.
| | - Dezeng Liu
- College of Life Science, Henan Normal University, Xinxiang, China
| |
Collapse
|
12
|
Molina MD, Cebrià F. Decoding Stem Cells: An Overview on Planarian Stem Cell Heterogeneity and Lineage Progression. Biomolecules 2021; 11:1532. [PMID: 34680165 PMCID: PMC8533874 DOI: 10.3390/biom11101532] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/26/2023] Open
Abstract
Planarians are flatworms capable of whole-body regeneration, able to regrow any missing body part after injury or amputation. The extraordinary regenerative capacity of planarians is based upon the presence in the adult of a large population of somatic pluripotent stem cells. These cells, called neoblasts, offer a unique system to study the process of stem cell specification and differentiation in vivo. In recent years, FACS-based isolation of neoblasts, RNAi functional analyses as well as high-throughput approaches such as single-cell sequencing have allowed a rapid progress in our understanding of many different aspects of neoblast biology. Here, we summarize our current knowledge on the molecular signatures that define planarian neoblasts heterogeneity, which includes a percentage of truly pluripotent stem cells, and guide the commitment of pluripotent neoblasts into lineage-specific progenitor cells, as well as their differentiation into specific planarian cell types.
Collapse
Affiliation(s)
- M. Dolores Molina
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), 08028 Barcelona, Spain
| | - Francesc Cebrià
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), 08028 Barcelona, Spain
| |
Collapse
|
13
|
Zheng H, Liu H, Xu Q, Wang W, Li L, Ye G, Wen X, Chen F, Yu Y. PI3K Plays an Essential Role in Planarian Regeneration and Tissue Maintenance. Front Cell Dev Biol 2021; 9:649656. [PMID: 34422792 PMCID: PMC8377419 DOI: 10.3389/fcell.2021.649656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K) signaling plays a central role in various biological processes, and its abnormality leads to a broad spectrum of human diseases, such as cancer, fibrosis, and immunological disorders. However, the mechanisms by which PI3K signaling regulates the behavior of stem cells during regeneration are poorly understood. Planarian flatworms possess abundant adult stem cells (called neoblasts) allowing them to develop remarkable regenerative capabilities, thus the animals represent an ideal model for studying stem cells and regenerative medicine in vivo. In this study, the spatiotemporal expression pattern of Djpi3k, a PI3K ortholog in the planarian Dugesia japonica, was investigated and suggests its potential role in wound response and tissue regeneration. A loss-of-function study was conducted using small molecules and RNA interference technique, providing evidence that PI3K signaling is required for blastema regrowth and cilia maintenance during planarian regeneration and homeostasis. Interestingly, the mitotic and apoptotic responses to amputation are substantially abated in PI3K inhibitor-treated regenerating animals, while knockdown of Djpi3k alleviates the mitotic response and postpones the peak of apoptotic cell death, which may contribute to the varying degrees of regenerative defects induced by the pharmacological and genetic approaches. These observations reveal novel roles for PI3K signaling in the regulation of the cellular responses to amputation during planarian regeneration and provide insights for investigating the disease-related genes in the regeneration-competent organism in vivo.
Collapse
Affiliation(s)
- Hanxue Zheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Hongbo Liu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Qian Xu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Wenjun Wang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Linfeng Li
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Gang Ye
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Xiaomin Wen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China.,Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yuan Yu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China.,Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| |
Collapse
|
14
|
Ceccarelli M, D'Andrea G, Micheli L, Gentile G, Cavallaro S, Merlino G, Papoff G, Tirone F. Tumor Growth in the High Frequency Medulloblastoma Mouse Model Ptch1 +/-/Tis21 KO Has a Specific Activation Signature of the PI3K/AKT/mTOR Pathway and Is Counteracted by the PI3K Inhibitor MEN1611. Front Oncol 2021; 11:692053. [PMID: 34395258 PMCID: PMC8362831 DOI: 10.3389/fonc.2021.692053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
We have previously generated a mouse model (Ptch1+/−/Tis21KO), which displays high frequency spontaneous medulloblastoma, a pediatric tumor of the cerebellum. Early postnatal cerebellar granule cell precursors (GCPs) of this model show, in consequence of the deletion of Tis21, a defect of the Cxcl3-dependent migration. We asked whether this migration defect, which forces GCPs to remain in the proliferative area at the cerebellar surface, would be the only inducer of their high frequency transformation. In this report we show, by further bioinformatic analysis of our microarray data of Ptch1+/−/Tis21KO GCPs, that, in addition to the migration defect, they show activation of the PI3K/AKT/mTOR pathway, as the mRNA levels of several activators of this pathway (e.g., Lars, Rraga, Dgkq, Pdgfd) are up-regulated, while some inhibitors (e.g. Smg1) are down-regulated. No such change is observed in the Ptch1+/− or Tis21KO background alone, indicating a peculiar synergy between these two genotypes. Thus we investigated, by mRNA and protein analysis, the role of PI3K/AKT/mTOR signaling in MBs and in nodules from primary Ptch1+/−/Tis21KO MB allografted in the flanks of immunosuppressed mice. Activation of the PI3K/AKT/mTOR pathway is seen in full-blown Ptch1+/−/Tis21KO MBs, relative to Ptch1+/−/Tis21WT MBs. In Ptch1+/−/Tis21KO MBs we observe that the proliferation of neoplastic GCPs increases while apoptosis decreases, in parallel with hyper-phosphorylation of the mTOR target S6, and, to a lower extent, of AKT. In nodules derived from primary Ptch1+/−/Tis21KO MBs, treatment with MEN1611, a novel PI3K inhibitor, causes a dramatic reduction of tumor growth, inhibiting proliferation and, conversely, increasing apoptosis, also of tumor CD15+ stem cells, responsible for long-term relapses. Additionally, the phosphorylation of AKT, S6 and 4EBP1 was significantly inhibited, indicating inactivation of the PI3K/AKT/mTOR pathway. Thus, PI3K/AKT/mTOR pathway activation contributes to Ptch1+/−/Tis21KO MB development and to high frequency tumorigenesis, observed when the Tis21 gene is down-regulated. MEN1611 could provide a promising therapy for MB, especially for patient with down-regulation of Btg2 (human ortholog of the murine Tis21 gene), which is frequently deregulated in Shh-type MBs.
Collapse
Affiliation(s)
- Manuela Ceccarelli
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| | - Giorgio D'Andrea
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| | - Laura Micheli
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| | - Giulia Gentile
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy
| | | | - Giuliana Papoff
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| | - Felice Tirone
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC-CNR), Rome, Italy
| |
Collapse
|
15
|
Gutiérrez-Gutiérrez Ó, Felix DA, Salvetti A, Amro EM, Thems A, Pietsch S, Koeberle A, Rudolph KL, González-Estévez C. Regeneration in starved planarians depends on TRiC/CCT subunits modulating the unfolded protein response. EMBO Rep 2021; 22:e52905. [PMID: 34190393 PMCID: PMC8344900 DOI: 10.15252/embr.202152905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/15/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Planarians are able to stand long periods of starvation by maintaining adult stem cell pools and regenerative capacity. The molecular pathways that are needed for the maintenance of regeneration during starvation are not known. Here, we show that down‐regulation of chaperonin TRiC/CCT subunits abrogates the regeneration capacity of planarians during starvation, but TRiC/CCT subunits are dispensable for regeneration in fed planarians. Under starvation, they are required to maintain mitotic fidelity and for blastema formation. We show that TRiC subunits modulate the unfolded protein response (UPR) and are required to maintain ATP levels in starved planarians. Regenerative defects in starved CCT‐depleted planarians can be rescued by either chemical induction of mild endoplasmic reticulum stress, which leads to induction of the UPR, or by the supplementation of fatty acids. Together, these results indicate that CCT‐dependent UPR induction promotes regeneration of planarians under food restriction.
Collapse
Affiliation(s)
| | - Daniel A Felix
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Alessandra Salvetti
- Department of Clinical and Experimental Medicine, Unit of Experimental Biology and Genetics, University of Pisa, Pisa, Italy
| | - Elias M Amro
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Anne Thems
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Stefan Pietsch
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Andreas Koeberle
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany.,Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - K Lenhard Rudolph
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | | |
Collapse
|
16
|
Molinaro AM, Lindsay‐Mosher N, Pearson BJ. Identification of TOR-responsive slow-cycling neoblasts in planarians. EMBO Rep 2021; 22:e50292. [PMID: 33511776 PMCID: PMC7926258 DOI: 10.15252/embr.202050292] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 11/19/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Epimorphic regeneration commonly relies on the activation of reserved stem cells to drive new cell production. The planarian Schmidtea mediterranea is among the best regenerators in nature, thanks to its large population of adult stem cells, called neoblasts. While neoblasts have long been known to drive regeneration, whether a subset of neoblasts is reserved for this purpose is unknown. Here, we revisit the idea of reserved neoblasts by approaching neoblast heterogeneity from a regulatory perspective. By implementing a new fluorescence-activated cell sorting strategy in planarians, we identify a population of neoblasts defined by low transcriptional activity. These RNAlow neoblasts are relatively slow-cycling at homeostasis and undergo a morphological regeneration response characterized by cell growth at 48 h post-amputation. At this time, RNAlow neoblasts proliferate in a TOR-dependent manner. Additionally, knockdown of the tumour suppressor Lrig-1, which is enriched in RNAlow neoblasts, results in RNAlow neoblast growth and hyperproliferation at homeostasis, and ultimately delays regeneration. We propose that slow-cycling RNAlow neoblasts represent a regeneration-reserved neoblast population.
Collapse
Affiliation(s)
- Alyssa M Molinaro
- Program in Developmental and Stem Cell BiologyHospital for Sick ChildrenTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
- Present address:
Department of Systems BiologyHarvard Medical SchoolBostonMAUSA
| | - Nicole Lindsay‐Mosher
- Program in Developmental and Stem Cell BiologyHospital for Sick ChildrenTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Bret J Pearson
- Program in Developmental and Stem Cell BiologyHospital for Sick ChildrenTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
- Ontario Institute for Cancer ResearchTorontoONCanada
| |
Collapse
|
17
|
Williams MC, Patel JH, Kakebeen AD, Wills AE. Nutrient availability contributes to a graded refractory period for regeneration in Xenopus tropicalis. Dev Biol 2021; 473:59-70. [PMID: 33484704 DOI: 10.1016/j.ydbio.2021.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/17/2020] [Accepted: 01/12/2021] [Indexed: 11/28/2022]
Abstract
Xenopus tadpoles are a unique model for regeneration in that they exhibit two distinct phases of age-specific regenerative competence. In Xenopus laevis, young tadpoles fully regenerate following major injuries such as tail transection, then transiently lose regenerative competence during the "refractory period" from stages 45-47. Regenerative competence is then regained in older tadpoles before being permanently lost during metamorphosis. Here we show that a similar refractory period exists in X. tropicalis. Notably, tadpoles lose regenerative competence gradually in X. tropicalis, with full regenerative competence lost at stage 47. We find that the refractory period coincides closely with depletion of maternal yolk stores and the onset of independent feeding, and so we hypothesized that it might be caused in part by nutrient stress. In support of this hypothesis, we find that cell proliferation declines throughout the tail as the refractory period approaches. When we block nutrient mobilization by inhibiting mTOR signaling, we find that tadpole growth and regeneration are reduced, while yolk stores persist. Finally, we are able to restore regenerative competence and cell proliferation during the refractory period by abundantly feeding tadpoles. Our study argues that nutrient stress contributes to lack of regenerative competence and introduces the X. tropicalis refractory period as a valuable new model for interrogating how metabolic constraints inform regeneration.
Collapse
Affiliation(s)
| | - Jeet H Patel
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Program in Molecular and Cellular Biology, University of Washington School of Medicine, Seattle, WA, USA
| | - Anneke D Kakebeen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Andrea E Wills
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
18
|
Deb S, Felix DA, Koch P, Deb MK, Szafranski K, Buder K, Sannai M, Groth M, Kirkpatrick J, Pietsch S, Gollowitzer A, Groß A, Riemenschneider P, Koeberle A, González‐Estévez C, Rudolph KL. Tnfaip2/exoc3-driven lipid metabolism is essential for stem cell differentiation and organ homeostasis. EMBO Rep 2021; 22:e49328. [PMID: 33300287 PMCID: PMC7788457 DOI: 10.15252/embr.201949328] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/21/2020] [Accepted: 10/29/2020] [Indexed: 01/07/2023] Open
Abstract
Lipid metabolism influences stem cell maintenance and differentiation but genetic factors that control these processes remain to be delineated. Here, we identify Tnfaip2 as an inhibitor of reprogramming of mouse fibroblasts into induced pluripotent stem cells. Tnfaip2 knockout impairs differentiation of embryonic stem cells (ESCs), and knockdown of the planarian para-ortholog, Smed-exoc3, abrogates in vivo tissue homeostasis and regeneration-processes that are driven by somatic stem cells. When stimulated to differentiate, Tnfaip2-deficient ESCs fail to induce synthesis of cellular triacylglycerol (TAG) and lipid droplets (LD) coinciding with reduced expression of vimentin (Vim)-a known inducer of LD formation. Smed-exoc3 depletion also causes a strong reduction of TAGs in planarians. The study shows that Tnfaip2 acts epistatically with and upstream of Vim in impairing cellular reprogramming. Supplementing palmitic acid (PA) and palmitoyl-L-carnitine (the mobilized form of PA) restores the differentiation capacity of Tnfaip2-deficient ESCs and organ maintenance in Smed-exoc3-depleted planarians. Together, these results identify a novel role of Tnfaip2 and exoc3 in controlling lipid metabolism, which is essential for ESC differentiation and planarian organ maintenance.
Collapse
Affiliation(s)
- Sarmistha Deb
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Daniel A Felix
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Philipp Koch
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | | | - Karol Szafranski
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Katrin Buder
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Mara Sannai
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - Marco Groth
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | | | - Stefan Pietsch
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
| | - André Gollowitzer
- Institute of PharmacyFriedrich‐Schiller‐UniversityJenaGermany
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Alexander Groß
- Institute of Medical Systems BiologyUlm UniversityUlmGermany
| | | | - Andreas Koeberle
- Institute of PharmacyFriedrich‐Schiller‐UniversityJenaGermany
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | | | - Karl Lenhard Rudolph
- Leibniz Institute on Aging – Fritz Lipmann Institute e.V.JenaGermany
- University Hospital JenaFriedrich Schiller UniversityJenaGermany
| |
Collapse
|
19
|
Song Q, Liu H, Zhen H, Zhao B. Autophagy and its role in regeneration and remodeling within invertebrate. Cell Biosci 2020; 10:111. [PMID: 32974004 PMCID: PMC7507827 DOI: 10.1186/s13578-020-00467-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/30/2020] [Indexed: 12/12/2022] Open
Abstract
Background Acting as a cellular cleaner by packaging and transporting defective proteins and organelles to lysosomes for breakdown, autophagic process is involved in the regulation of cell remodeling after cell damage or cell death in both vertebrate and invertebrate. In human, limitations on the regenerative capacity of specific tissues and organs make it difficult to recover from diseases. Comprehensive understanding on its mechanism within invertebrate have strong potential provide helpful information for challenging these diseases. Method In this study, recent findings on the autophagy function in three invertebrates including planarian, hydra and leech with remarkable regenerative ability were summarized. Furthermore, molecular phylogenetic analyses of DjATGs and HvATGs were performed on these three invertebrates compared to that of Saccharomyces cerevisiae, Caenorhabditis elegans, Drosophila melanogaster, Mus musculus and Homo sapiens. Results In comparison with Scerevisiae, C elegans, D melanogaster, M musculus and human, our analysis exhibits the following characteristics of autophagy and its function in regeneration within invertebrate. Phylogenetical analysis of ATGs revealed that most autophagy-related genes (ATGs) were highly similar to their homologs in other species, which indicates that autophagy is a highly conservative biological function in both vertebrate and invertebrate. Structurally, almost all the core amino acids necessary for the function of ATG8 in mammal were observed in invertebrate HvATG8s and DjATG8s. For instance, ubiquitin-like domain as a signature structure in each ATG8, was observed in all ATG8s in three invertebrates. Basically, autophagy plays a key role in the regulation of regeneration in planarian. DjATG8-2 and DjATG8-3 associated with mTOR signaling pathway are sophisticated in the invertebrate tissue/organ regeneration. Furthermore, autophagy is involved in the pathway of neutralization of toxic molecules input from blood digestion in the leech. Conclusions The recent investigations on autophagy in invertebrate including planarian, hydra and leech suggest that autophagy is evolutionally conserved from yeast to mammals. The fundamental role of its biological function in the invertebrate contributing to the regeneration and maintenance of cellular homeostasis in these three organisms could make tremendous information to confront life threatening diseases in human including cancers and cardiac disorders.
Collapse
Affiliation(s)
- Qian Song
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Zibo, 255049 Shandong China
| | - Hongjin Liu
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Zibo, 255049 Shandong China
| | - Hui Zhen
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Zibo, 255049 Shandong China
| | - Bosheng Zhao
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Zibo, 255049 Shandong China
| |
Collapse
|
20
|
Zeng S, Liu S, Feng J, Gao J, Xue F. MicroRNA-32 promotes ovarian cancer cell proliferation and motility by targeting SMG1. Oncol Lett 2020; 20:733-741. [PMID: 32565999 PMCID: PMC7285996 DOI: 10.3892/ol.2020.11624] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 11/22/2019] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy and one of the leading causes of cancer-related deaths among women. Metastasis is the main cause of poor prognosis in OC. MicroRNA (miRNA/miR) has been shown to play an important role in tumorigenesis and metastasis in various cancer types by affecting the expression of its targets. In the present study, the role of miR-32 (miR-32-5p) in OC was explored. Reverse transcription-quantitative PCR results showed that miR-32 expression was significantly upregulated in both OC tissues and cell lines. Inhibition of miR-32 by transfection with miR-32 inhibitor in OC cells markedly suppressed cell proliferation, migration and invasion. In addition, a luciferase assay showed that suppressor of morphogenesis in genitalia 1 (SMG1) is a direct target of miR-32, and interference in SMG1 expression with transfection of SMG1 small hairpin RNA restored miR-32-mediated OC cell proliferation, migration and invasion. Taken together, these results indicate that miR-32 may promote OC cell growth and motility by targeting SMG1. The data of the present study suggest that miR-32 may serve as a potential therapeutic target for OC treatment in the future.
Collapse
Affiliation(s)
- Saitian Zeng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Department of Gynecology and Obstetrics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Shikai Liu
- Department of Gynecology and Obstetrics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Jing Feng
- Department of Gynecology and Obstetrics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Jiefan Gao
- Department of Gynecology and Obstetrics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, P.R. China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
21
|
Ziman B, Karabinis P, Barghouth P, Oviedo NJ. Sirtuin-1 regulates organismal growth by altering feeding behavior and intestinal morphology in planarians. J Cell Sci 2020; 133:jcs239467. [PMID: 32265271 PMCID: PMC7272345 DOI: 10.1242/jcs.239467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 03/19/2020] [Indexed: 01/03/2023] Open
Abstract
Nutrient availability upon feeding leads to an increase in body size in the planarian Schmidtea mediterranea However, it remains unclear how food consumption integrates with cell division at the organismal level. Here, we show that the NAD-dependent protein deacetylases sirtuins are evolutionarily conserved in planarians, and specifically demonstrate that the homolog of human sirtuin-1 (SIRT1) (encoded by Smed-Sirt-1), regulates organismal growth by impairing both feeding behavior and intestinal morphology. Disruption of Smed-Sirt-1 with RNAi or pharmacological inhibition of Sirtuin-1 leads to reduced animal growth. Conversely, enhancement of Sirtuin-1 activity with resveratrol accelerates growth. Differences in growth rates were associated with changes in the amount of time taken to locate food and overall food consumption. Furthermore, Smed-Sirt-1(RNAi) animals displayed reduced cell death and increased stem cell proliferation accompanied by impaired expression of intestinal lineage progenitors and reduced branching of the gut. Taken together, our findings indicate that Sirtuin-1 is a crucial metabolic hub capable of controlling animal behavior, tissue renewal and morphogenesis of the adult intestine.
Collapse
Affiliation(s)
- Benjamin Ziman
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Peter Karabinis
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Paul Barghouth
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
| | - Néstor J Oviedo
- Department of Molecular and Cell Biology, University of California, Merced, CA 95343, USA
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA 95343, USA
- Health Sciences Research Institute, University of California, Merced, CA 95343, USA
| |
Collapse
|
22
|
Wouters A, Ploem JP, Langie SAS, Artois T, Aboobaker A, Smeets K. Regenerative responses following DNA damage - β-catenin mediates head regrowth in the planarian Schmidtea mediterranea. J Cell Sci 2020; 133:jcs237545. [PMID: 32107291 DOI: 10.1242/jcs.237545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 02/09/2020] [Indexed: 12/12/2022] Open
Abstract
Pluripotent stem cells hold great potential for regenerative medicine. Increased replication and division, such is the case during regeneration, concomitantly increases the risk of adverse outcomes through the acquisition of mutations. Seeking for driving mechanisms of such outcomes, we challenged a pluripotent stem cell system during the tightly controlled regeneration process in the planarian Schmidtea mediterranea Exposure to the genotoxic compound methyl methanesulfonate (MMS) revealed that despite a similar DNA-damaging effect along the anteroposterior axis of intact animals, responses differed between anterior and posterior fragments after amputation. Stem cell proliferation and differentiation proceeded successfully in the amputated heads, leading to regeneration of missing tissues. Stem cells in the amputated tails showed decreased proliferation and differentiation capacity. As a result, tails could not regenerate. Interference with the body-axis-associated component β-catenin-1 increased regenerative success in tail fragments by stimulating proliferation at an early time point. Our results suggest that differences in the Wnt signalling gradient along the body axis modulate stem cell responses to MMS.
Collapse
Affiliation(s)
- Annelies Wouters
- Zoology, Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Jan-Pieter Ploem
- Zoology, Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Sabine A S Langie
- Vito Health, 2400 Mol, Belgium
- Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Tom Artois
- Zoology, Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Aziz Aboobaker
- Department of Zoology, University of Oxford, Oxford OX1 3PS, UK
| | - Karen Smeets
- Zoology, Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| |
Collapse
|
23
|
Pascual-Carreras E, Marin-Barba M, Herrera-Úbeda C, Font-Martín D, Eckelt K, de Sousa N, García-Fernández J, Saló E, Adell T. Planarian cell number depends on blitzschnell, a novel gene family that balances cell proliferation and cell death. Development 2020; 147:dev.184044. [PMID: 32122990 DOI: 10.1242/dev.184044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/19/2020] [Indexed: 01/14/2023]
Abstract
Control of cell number is crucial to define body size during animal development and to restrict tumoral transformation. The cell number is determined by the balance between cell proliferation and cell death. Although many genes are known to regulate those processes, the molecular mechanisms underlying the relationship between cell number and body size remain poorly understood. This relationship can be better understood by studying planarians, flatworms that continuously change their body size according to nutrient availability. We identified a novel gene family, blitzschnell (bls), that consists of de novo and taxonomically restricted genes that control cell proliferation:cell death ratio. Their silencing promotes faster regeneration and increases cell number during homeostasis. Importantly, this increase in cell number leads to an increase in body size only in a nutrient-rich environment; in starved planarians, silencing results in a decrease in cell size and cell accumulation that ultimately produces overgrowths. bls expression is downregulated after feeding and is related to activity of the insulin/Akt/mTOR network, suggesting that the bls family evolved in planarians as an additional mechanism for restricting cell number in nutrient-fluctuating environments.
Collapse
Affiliation(s)
- Eudald Pascual-Carreras
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine, Universitat de Barcelona, Barcelona 08028, Catalunya, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Catalunya, Spain
| | - Marta Marin-Barba
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Carlos Herrera-Úbeda
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine, Universitat de Barcelona, Barcelona 08028, Catalunya, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Catalunya, Spain
| | - Daniel Font-Martín
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine, Universitat de Barcelona, Barcelona 08028, Catalunya, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Catalunya, Spain
| | - Kay Eckelt
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine, Universitat de Barcelona, Barcelona 08028, Catalunya, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Catalunya, Spain
| | - Nidia de Sousa
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine, Universitat de Barcelona, Barcelona 08028, Catalunya, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Catalunya, Spain
| | - Jordi García-Fernández
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine, Universitat de Barcelona, Barcelona 08028, Catalunya, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Catalunya, Spain
| | - Emili Saló
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine, Universitat de Barcelona, Barcelona 08028, Catalunya, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Catalunya, Spain
| | - Teresa Adell
- Department of Genetics, Microbiology and Statistics and Institute of Biomedicine, Universitat de Barcelona, Barcelona 08028, Catalunya, Spain .,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Catalunya, Spain
| |
Collapse
|
24
|
González-Estévez C, Flores I. Fasting for stem cell rejuvenation. Aging (Albany NY) 2020; 12:4048-4049. [PMID: 32160590 PMCID: PMC7093158 DOI: 10.18632/aging.102912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/05/2020] [Indexed: 11/25/2022]
Affiliation(s)
| | - Ignacio Flores
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid 28029, Spain.,Centro de Biologia Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain
| |
Collapse
|
25
|
Allen EA, Baehrecke EH. Autophagy in animal development. Cell Death Differ 2020; 27:903-918. [PMID: 31988494 PMCID: PMC7206001 DOI: 10.1038/s41418-020-0497-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/19/2019] [Accepted: 01/07/2020] [Indexed: 01/13/2023] Open
Abstract
Macroautophagy (autophagy) delivers intracellular constituents to the lysosome to promote catabolism. During development in multiple organisms, autophagy mediates various cellular processes, including survival during starvation, programmed cell death, phagocytosis, organelle elimination, and miRNA regulation. Our current understanding of autophagy has been enhanced by developmental biology research during the last quarter of a century. Through experiments that focus on animal development, fundamental mechanisms that control autophagy and that contribute to disease were elucidated. Studies in embryos revealed specific autophagy molecules that mediate the removal of paternally derived mitochondria, and identified autophagy components that clear protein aggregates during development. Importantly, defects in mtDNA inheritance, or removal of paternal mtDNA via mitochondrial autophagy, can contribute to mitochondrial-associated disease. In addition, impairment of the clearance of protein aggregates by autophagy underlies neurodegenerative diseases. Experiments in multiple organisms also reveal conserved mechanisms of tissue remodeling that rely on the cooperation between autophagy and apoptosis to clear cell corpses, and defects in autophagy and apoptotic cell clearance can contribute to inflammation and autoimmunity. Here we provide an overview of key developmental processes that are mediated by autophagy in multiple animals.
Collapse
Affiliation(s)
- Elizabeth A Allen
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 423 Lazare Research Building, 364 Plantation St., Worcester, MA, 01655, USA
| | - Eric H Baehrecke
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 423 Lazare Research Building, 364 Plantation St., Worcester, MA, 01655, USA.
| |
Collapse
|
26
|
Kang J, Dong Z, Hao Q, Wang J, Chen G, Liu D. The regulation of rapamycin in planarian Dugesia japonica Ichikawa & Kawakatsu, 1964 regeneration according to TOR signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 185:109680. [PMID: 31546204 DOI: 10.1016/j.ecoenv.2019.109680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 06/10/2023]
Abstract
The freshwater planarian mostly lives in the upper reaches of springs and rivers. Generally, it is realized as a suitable warning indicator of environmental toxicants. The freshwater planarian Dugesia japonica has a powerful regenerative capability and can regenerate a new individual including a complete central nervous system in one week. Rapamycin is an inhibitor of mammalian TORC1 (target of rapamycin complex-1) and used in the treatment of some diseases like cancer, cardiovascular and neurological diseases. However, the roles of rapamycin in the regulation of planarian regeneration remain to be elucidated. In present study, freshwater planarians D. japonica were firstly treated with 1 μM rapamycin for 18 h exposures and the expression patterns of Djtor was analyzed by the whole-mount in situ hybridization (WISH). Our results indicated rapamycin could strongly inhibit Djtor expression in planarian D. japonica and cause asymmetric blastemas and neuronal defects in planarians. Furthermore, knockdown of Djtor gene in planarians using RNA interference resulted in the suppression of downstream autophagy genes. These findings suggested that rapamycin might regulate freshwater planarian regeneration via Djtor signaling pathway.
Collapse
Affiliation(s)
- Jing Kang
- College of Life Science, Henan Normal University, Xinxiang, China; College of Life Science, Xingxiang Medical University, Xinxiang, China.
| | - Zimei Dong
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Qin Hao
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Jing Wang
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Guangwen Chen
- College of Life Science, Henan Normal University, Xinxiang, China.
| | - Dezeng Liu
- College of Life Science, Henan Normal University, Xinxiang, China
| |
Collapse
|
27
|
Mai S, Xiao R, Shi L, Zhou X, Yang T, Zhang M, Weng N, Zhao X, Wang R, Liu J, Sun R, Qin H, Wang H. MicroRNA-18a promotes cancer progression through SMG1 suppression and mTOR pathway activation in nasopharyngeal carcinoma. Cell Death Dis 2019; 10:819. [PMID: 31659158 PMCID: PMC6817863 DOI: 10.1038/s41419-019-2060-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/17/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
miR-18a has been reported to be upregulated in nasopharyngeal carcinoma (NPC) tissues by microarray assays. However, the roles and the underlying mechanisms of miR-18a in NPC remain poorly understood. Here we demonstrated by real-time RT-PCR that miR-18a expression is upregulated in NPC tissues, and positively correlated with tumor size and TNM stage. Moreover, miR-18a expression could be upregulated by NF-κB activation or Epstein-Barr virus encoded latent membrane protein 1 expression. The ectopic expression of miR-18a promoted NPC cell proliferation, migration and invasion, while the repression of miR-18a had opposite effects. Candidate genes under regulation by miR-18a were screened out through a whole-genome microarray assay, further identified by a reporter assay and verified in clinical samples. SMG1, a member of the phosphoinositide 3-kinase-related kinases family and an mTOR antagonist, was identified as functional target of miR-18a. Our results confirmed that miR-18a exerts its oncogenic role through suppression of SMG1 and activation of mTOR pathway in NPC cells. Importantly, in vivo xenograft tumor growth in nude mice was effectively inhibited by intratumor injection of miR-18a antagomir. Our data support an oncogenic role of miR-18a through a novel miR-18a/SMG1/mTOR axis and suggest that the antitumor effects of antagomir-18a may make it suitable for NPC therapy.
Collapse
Affiliation(s)
- ShiJuan Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - RuoWen Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lu Shi
- Department of thoracic oncology, the cancer center of the fifth affiliated hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - XiaoMin Zhou
- ZhouKou Hospital of Traditional Chinese Medicine, Zhoukou, 466000, China
| | - Te Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - MeiYin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - NuoQing Weng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - XinGe Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - RuiQi Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ji Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - HaiDe Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - HuiYun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
28
|
Yue Z, Lv Z, Shao Y, Zhang W, Zhao X, Guo M, Li C. Cloning and characterization of the target protein subunit lst8 of rapamycin in Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2019; 92:460-468. [PMID: 31233778 DOI: 10.1016/j.fsi.2019.06.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 06/09/2023]
Abstract
Autophagy plays an important role in the immune defense systems of vertebrates through the interaction between the lethal with SEC13 protein 8 (lst8) and the mechanistic target of rapamycin. In the present study, a novel invertebrate lst8 homologue is identified from Apostichopus japonicus (designated as Ajlst8) via polymerase chain reaction. The full-length complementary DNA of Ajlst8 comprises a 5'-untranslated region (UTR) of 78 base pair (bp), a 3'-UTR of 479 bp, and a putative open reading frame of 951 bp; hence, 316 amino acids are encoded. Structural analysis shows that the deduced amino acid of Ajlst8 shares six typical WD40 domains (28 aa-248 aa). Spatial expression analysis indicates that Ajlst8 is ubiquitously expressed in all the examined tissues, with a larger magnitude in coelomocytes. Vibrio splendidus infection in vivo and lipopolysaccharide exposure in vitro can significantly upregulate the messenger RNA expression of Ajlst8 by 2.39-fold and 1.93-fold compared with the control group, respectively. LPS exposure could also significantly induced the protein level of Ajlst8 to 2.38-fold and the autophagy level was markedly increased by 3.08-fold under same condition. The RNA interference of Ajlst8 in primary coelomocytes also reduces the relative expression of autophagy with a 0.71-fold decrease in the ratio of LC3-II/LC3-I compared with that in the control group. These results indicate that Ajlst8 is a novel immune regulator that may be involved in the antibacterial response process of sea cucumber by regulating autophagy.
Collapse
Affiliation(s)
- Zongxu Yue
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Zhimeng Lv
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Yina Shao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Weiwei Zhang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Xuelin Zhao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Ming Guo
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
29
|
Iglesias M, Felix DA, Gutiérrez-Gutiérrez Ó, De Miguel-Bonet MDM, Sahu S, Fernández-Varas B, Perona R, Aboobaker AA, Flores I, González-Estévez C. Downregulation of mTOR Signaling Increases Stem Cell Population Telomere Length during Starvation of Immortal Planarians. Stem Cell Reports 2019; 13:405-418. [PMID: 31353226 PMCID: PMC6700675 DOI: 10.1016/j.stemcr.2019.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 12/20/2022] Open
Abstract
Reduction of caloric intake delays and prevents age-associated diseases and extends the life span in many organisms. It may be that these benefits are due to positive effects of caloric restriction on stem cell function. We use the planarian model Schmidtea mediterranea, an immortal animal that adapts to long periods of starvation by shrinking in size, to investigate the effects of starvation on telomere length. We show that the longest telomeres are a general signature of planarian adult stem cells. We also observe that starvation leads to an enrichment of stem cells with the longest telomeres and that this enrichment is dependent on mTOR signaling. We propose that one important effect of starvation for the rejuvenation of the adult stem cell pool is through increasing the median telomere length in somatic stem cells. Such a mechanism has broad implications for how dietary effects on aging are mediated at the whole-organism level.
Collapse
Affiliation(s)
- Marta Iglesias
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Daniel A Felix
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany
| | | | - Maria Del Mar De Miguel-Bonet
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Sounak Sahu
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| | - Beatriz Fernández-Varas
- Instituto de Investigaciones Biomédicas CSIC/UAM, IDiPaz, Arturo Duperier 4, 28029 Madrid, Spain
| | - Rosario Perona
- Instituto de Investigaciones Biomédicas CSIC/UAM, IDiPaz, Arturo Duperier 4, 28029 Madrid, Spain; Ciber Network on Rare Diseases (CIBERER), C/ Alvaro de Bazan, 10, 46010 Valencia, Spain
| | - A Aziz Aboobaker
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| | - Ignacio Flores
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| | - Cristina González-Estévez
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| |
Collapse
|
30
|
Cote LE, Simental E, Reddien PW. Muscle functions as a connective tissue and source of extracellular matrix in planarians. Nat Commun 2019; 10:1592. [PMID: 30962434 PMCID: PMC6453901 DOI: 10.1038/s41467-019-09539-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/15/2019] [Indexed: 12/24/2022] Open
Abstract
Regeneration and tissue turnover require new cell production and positional information. Planarians are flatworms capable of regenerating all body parts using a population of stem cells called neoblasts. The positional information required for tissue patterning is primarily harbored by muscle cells, which also control body contraction. Here we produce an in silico planarian matrisome and use recent whole-animal single-cell-transcriptome data to determine that muscle is a major source of extracellular matrix (ECM). No other ECM-secreting, fibroblast-like cell type was detected. Instead, muscle cells express core ECM components, including all 19 collagen-encoding genes. Inhibition of muscle-expressed hemicentin-1 (hmcn-1), which encodes a highly conserved ECM glycoprotein, results in ectopic peripheral localization of cells, including neoblasts, outside of the muscle layer. ECM secretion and hmcn-1-dependent maintenance of tissue separation indicate that muscle functions as a planarian connective tissue, raising the possibility of broad roles for connective tissue in adult positional information. How the cellular source of positional information compares across regenerative animals is unclear. Here, the authors find that planarian muscle, which harbours positional information, acts as a connective tissue by being a major site of matrisome gene expression and by maintaining tissue architecture.
Collapse
Affiliation(s)
- Lauren E Cote
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 455 Main St, Cambridge, MA, 02142, USA
| | - Eric Simental
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 455 Main St, Cambridge, MA, 02142, USA.,University of California San Francisco, 600 16th Street, San Francisco, CA, 94143, USA
| | - Peter W Reddien
- Howard Hughes Medical Institute, Whitehead Institute, and Department of Biology, Massachusetts Institute of Technology, 455 Main St, Cambridge, MA, 02142, USA.
| |
Collapse
|
31
|
Zhou JY, Huang DG, Qin YC, Li XG, Gao CQ, Yan HC, Wang XQ. mTORC1 signaling activation increases intestinal stem cell activity and promotes epithelial cell proliferation. J Cell Physiol 2019; 234:19028-19038. [PMID: 30937902 DOI: 10.1002/jcp.28542] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
Abstract
The crypt-villus axis of the intestine undergoes a continuous renewal process that is driven by intestinal stem cells (ISCs). However, the homeostasis is disturbed under constant exposure to high ambient temperatures, and the precise mechanism is unclear. We found that both EdU+ and Ki67+ cell ratios were significantly reduced after exposure to 41°C, as well as the protein synthesis rate of IPEC-J2 cells, and the expression of ubiquitin and heat shock protein 60, 70, and 90 were significantly increased. Additionally, heat exposure decreased enteroid expansion and budding efficiency, as well as induced apoptosis after 48 hr; however, no significant difference was observed in the apoptosis ratio after 24 hr. In the process of heat exposure, the mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway was significantly inhibited in both IPEC-J2 cells and enteroids. Correspondingly, treatment of IPEC-J2 and enteroids with the mTORC1 agonist MHY1485 at 41°C significantly attenuated the inhibition of proliferation and protein synthesis, increased the ISC activity, and promoted expansion and budding of enteroid. In summary, we conclude that the mTORC1 signaling pathway regulates intestinal epithelial cell and stem cell activity during heat exposure-induced injury.
Collapse
Affiliation(s)
- Jia-Yi Zhou
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Deng-Gui Huang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ying-Chao Qin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiang-Guang Li
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Chun-Qi Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Hui-Chao Yan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiu-Qi Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
32
|
Stevens AS, Wouters A, Ploem JP, Pirotte N, Van Roten A, Willems M, Hellings N, Franken C, Koppen G, Artois T, Plusquin M, Smeets K. Planarians Customize Their Stem Cell Responses Following Genotoxic Stress as a Function of Exposure Time and Regenerative State. Toxicol Sci 2019; 162:251-263. [PMID: 29145667 DOI: 10.1093/toxsci/kfx247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aiming to in vivo characterize the responses of pluripotent stem cells and regenerative tissues to carcinogenic stress, we employed the highly regenerative organism Schmidtea mediterranea. Its broad regenerative capacities are attributable to a large pool of pluripotent stem cells, which are considered key players in the lower vulnerability toward chemically induced carcinogenesis observed in regenerative organisms. Schmidtea mediterranea is, therefore, an ideal model to study pluripotent stem cell responses with stem cells residing in their natural environment. Including microenvironmental alterations is important, as the surrounding niche influences the onset of oncogenic events. Both short- (3 days) and long-term (17 days) exposures to the genotoxic carcinogen methyl methanesulfonate (50 µM) were evaluated during homeostasis and animal regeneration, two situations that render altered cellular niches. In both cases, MMS-induced DNA damage was observed, which provoked a decrease in proliferation on the short term. The outcome of DNA damage responses following long-term exposure differed between homeostatic and regenerating animals. During regeneration, DNA repair systems were more easily activated than in animals in homeostasis, where apoptosis was an important outcome. Knockdown experiments confirmed the importance of DNA repair systems during carcinogenic exposure in regenerating animals as knockdown of rad51 induced a stem cell-depleted phenotype, after regeneration was completed.
Collapse
Affiliation(s)
- An-Sofie Stevens
- Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Annelies Wouters
- Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Jan-Pieter Ploem
- Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Nicky Pirotte
- Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Andromeda Van Roten
- Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Maxime Willems
- Laboratory of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium.,Laboratory of Environmental Toxicology & Aquatic Ecology, Ghent University, 9000 Ghent, Belgium
| | - Niels Hellings
- Biomedical Research Institute, Hasselt University, 3590 Diepenbeek, Belgium
| | - Carmen Franken
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), 2400 Mol, Belgium
| | - Gudrun Koppen
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), 2400 Mol, Belgium
| | - Tom Artois
- Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Michelle Plusquin
- Environmental Biology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Karen Smeets
- Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| |
Collapse
|
33
|
Tunes LG, Allen JM, Zayas RM, do Monte-Neto RL. Planarians as models to investigate the bioactivity of gold(I) complexes in vivo. Sci Rep 2018; 8:16180. [PMID: 30385794 PMCID: PMC6212559 DOI: 10.1038/s41598-018-34558-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/18/2018] [Indexed: 02/02/2023] Open
Abstract
Gold(I)-containing complexes are used in drug discovery research for rheumatoid arthritis, cancer, and parasitic infections. In this study, we tested the bioactivity of gold(I) complexes in vivo using planarians. The planarian Schmidtea mediterranea possesses orthologues of tumor suppressor genes, such as p53, that, when silenced, cause deregulation of cell proliferation and apoptosis. In this context, we tested two triethylphosphine-gold(I) complexes (AdO and AdT) to determine if they can attenuate phenotypes that result from p53 inhibition. First, we identified the drug concentration that did not affect survival or regeneration and evaluated the drug's effect on cell division and apoptosis. We found that AdT treatment decreased the number of mitotic cells and that all drug treatments increased the number of apoptotic cells. We then performed p53(RNAi) and drug treatments concomitantly and observed the phenotype progression. Drug treatment increased survival three-fold and decreased apoptosis, which resulted in an attenuated phenotype. Our results indicate that planarians can be treated with gold(I) complexes, and that this treatment can diminish the p53(RNAi) phenotype and extend survival. In this work we show that planarians can be used as a model to study the in vivo effect of gold(I) complexes and to further investigate their mechanisms of action.
Collapse
Affiliation(s)
- Luiza G Tunes
- Instituto René Rachou - Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
- Department of Biology, San Diego State University, San Diego, California, USA
| | - John M Allen
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Ricardo M Zayas
- Department of Biology, San Diego State University, San Diego, California, USA.
| | | |
Collapse
|
34
|
Mihaylova Y, Abnave P, Kao D, Hughes S, Lai A, Jaber-Hijazi F, Kosaka N, Aboobaker AA. Conservation of epigenetic regulation by the MLL3/4 tumour suppressor in planarian pluripotent stem cells. Nat Commun 2018; 9:3633. [PMID: 30194301 PMCID: PMC6128892 DOI: 10.1038/s41467-018-06092-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/13/2018] [Indexed: 12/18/2022] Open
Abstract
Currently, little is known about the evolution of epigenetic regulation in animal stem cells. Here we demonstrate, using the planarian stem cell system to investigate the role of the COMPASS family of MLL3/4 histone methyltransferases that their function as tumor suppressors in mammalian stem cells is conserved over a long evolutionary distance. To investigate the potential conservation of a genome-wide epigenetic regulatory program in animal stem cells, we assess the effects of Mll3/4 loss of function by performing RNA-seq and ChIP-seq on the G2/M planarian stem cell population, part of which contributes to the formation of outgrowths. We find many oncogenes and tumor suppressors among the affected genes that are likely candidates for mediating MLL3/4 tumor suppression function. Our work demonstrates conservation of an important epigenetic regulatory program in animals and highlights the utility of the planarian model system for studying epigenetic regulation.
Collapse
Affiliation(s)
- Yuliana Mihaylova
- Department of Zoology, Tinbergen Building, South Parks Road, Oxford, OX1 3PS, UK
| | - Prasad Abnave
- Department of Zoology, Tinbergen Building, South Parks Road, Oxford, OX1 3PS, UK
| | - Damian Kao
- Department of Zoology, Tinbergen Building, South Parks Road, Oxford, OX1 3PS, UK
| | - Samantha Hughes
- HAN University of Applied Sciences, Institute of Applied Sciences, Laan van Scheut 2, 6525EM, Nijmegen, The Netherlands
| | - Alvina Lai
- Department of Zoology, Tinbergen Building, South Parks Road, Oxford, OX1 3PS, UK
| | - Farah Jaber-Hijazi
- Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - Nobuyoshi Kosaka
- Department of Zoology, Tinbergen Building, South Parks Road, Oxford, OX1 3PS, UK
| | - A Aziz Aboobaker
- Department of Zoology, Tinbergen Building, South Parks Road, Oxford, OX1 3PS, UK.
| |
Collapse
|
35
|
Van Roten A, Barakat AZAZ, Wouters A, Tran TA, Mouton S, Noben JP, Gentile L, Smeets K. A carcinogenic trigger to study the function of tumor suppressor genes in Schmidtea mediterranea. Dis Model Mech 2018; 11:dmm032573. [PMID: 29967069 PMCID: PMC6176991 DOI: 10.1242/dmm.032573] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 06/25/2018] [Indexed: 12/30/2022] Open
Abstract
Planarians have been long known for their regenerative ability, which hinges on pluripotency. Recently, however, the planarian model has been successfully established for routine toxicological screens aimed to assess overproliferation, mutagenicity and tumorigenesis. In this study, we focused on planarian tumor suppressor genes (TSGs) and their role during chemically induced carcinogenic stress in Schmidtea mediterranea Combining in silico and proteomic screens with exposure to human carcinogen type 1A agent cadmium (Cd), we showed that many TSGs have a function in stem cells and that, in general, exposure to Cd accelerated the onset and increased the severity of the observed phenotype. This suggested that the interaction between environmental and genetic factors plays an important role in tumor development in S. mediterranea Therefore, we further focused on the synergistic effects of Cd exposure and p53 knockdown (KD) at the cellular and molecular levels. Cd also produced a specific proteomic landscape in homeostatic animals, with 172 proteins differentially expressed, 43 of which were downregulated. Several of these proteins have tumor suppressor function in human and other animals, namely Wilms Tumor 1 Associated Protein (WT1), Heat Shock Protein 90 (HSP90), Glioma Pathogenesis-Related Protein 1 (GLIPR1) and Matrix Metalloproteinase B (Smed-MMPB). Both Glipr1 and MmpB KD produced large outgrowths, epidermal lesions and epidermal blisters. The epidermal blisters that formed as a consequence of Smed-MmpB KD were populated by smedwi1+ cells, many of which were actively proliferating, while large outgrowths contained ectopically differentiated structures, such as photoreceptors, nervous tissue and a small pharynx. In conclusion, Smed-MmpB is a planarian TSG that prevents stem cell proliferation and differentiation outside the proper milieu.
Collapse
Affiliation(s)
- Andromeda Van Roten
- Zoology: Biodiversity and Toxicology, Hasselt University-Campus Diepenbeek, Agoralaan 1, Gebouw D, 3590, Diepenbeek, Belgium
| | - Amal Zohir Abo-Zeid Barakat
- Planarian Stem Cell Laboratory, Max Planck Institute for Molecular Biomedicine, von Esmarch-str. 54, 48149, Münster, Germany
| | - Annelies Wouters
- Zoology: Biodiversity and Toxicology, Hasselt University-Campus Diepenbeek, Agoralaan 1, Gebouw D, 3590 Diepenbeek, Belgium
| | - Thao Anh Tran
- Pluripotency and Regeneration Group, Fraunhofer Institute for Biomedical Engineering, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Stijn Mouton
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, 9713, Groningen, The Netherlands
| | - Jean-Paul Noben
- Biomedical Research Institute, Hasselt University and Transnationale Universiteit Limburg, School of Life Sciences, 3590, Diepenbeek, Belgium
| | - Luca Gentile
- Planarian Stem Cell Laboratory, Max Planck Institute for Molecular Biomedicine, von Esmarch-str. 54, 48149, Münster, Germany
| | - Karen Smeets
- Zoology: Biodiversity and Toxicology, Hasselt University-Campus Diepenbeek, Agoralaan 1, Gebouw D, 3590, Diepenbeek, Belgium
| |
Collapse
|
36
|
Felix DA, Gutiérrez-Gutiérrez Ó, Espada L, Thems A, González-Estévez C. It is not all about regeneration: Planarians striking power to stand starvation. Semin Cell Dev Biol 2018; 87:169-181. [PMID: 29705301 DOI: 10.1016/j.semcdb.2018.04.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/11/2018] [Accepted: 04/25/2018] [Indexed: 12/12/2022]
Abstract
All living forms, prokaryotes as eukaryotes, have some means of adaptation to food scarcity, which extends the survival chances under extreme environmental conditions. Nowadays we know that dietary interventions, including fasting, extends lifespan of many organisms and can also protect against age-related diseases including in humans. Therefore, the capacity of adapting to periods of food scarcity may have evolved billions of years ago not only to allow immediate organismal survival but also to be able to extend organismal lifespan or at least to lead to a healthier remaining lifespan. Planarians have been the center of attention since more than two centuries because of their astonishing power of full body regeneration that relies on a large amount of adult stem cells or neoblasts. However, they also present an often-overlooked characteristic. They are able to stand long time starvation. Planarians have adapted to periods of fasting by shrinking or degrowing. Here we will review the published data about starvation in planarians and conclude with the possibility of starvation being one of the processes that rejuvenate the planarian, thus explaining the historical notion of non-ageing planarians.
Collapse
Affiliation(s)
- Daniel A Felix
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Óscar Gutiérrez-Gutiérrez
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Lilia Espada
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Anne Thems
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| | - Cristina González-Estévez
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany.
| |
Collapse
|
37
|
DNA damage and tissue repair: What we can learn from planaria. Semin Cell Dev Biol 2018; 87:145-159. [PMID: 29727725 DOI: 10.1016/j.semcdb.2018.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/22/2018] [Accepted: 04/30/2018] [Indexed: 12/21/2022]
Abstract
Faithful renewal of aging and damaged tissues is central to organismal lifespan. Stem cells (SCs) generate the cellular progeny that replenish adult tissues across the body but this task becomes increasingly compromised over time. The age related decline in SC-mediated tissue maintenance is a multifactorial event that commonly affects genome integrity. The presence of DNA damage in SCs that are under continuous demand to divide poses a great risk for age-related disorders such as cancer. However, performing analysis of SCs with genomic instability and the DNA damage response during tissue renewal present significant challenges. Here we introduce an alternative experimental system based on the planaria flatworm Schmidtea mediterranea to address at the organismal level studies intersecting SC-mediated tissue renewal in the presence of genomic instability. Planaria have abundant SCs (neoblasts) that maintain high rates of cellular turnover and a variety of molecular tools have been developed to induce DNA damage and dissect how neoblasts respond to this stressor. S. mediterranea displays high evolutionary conservation of DNA repair mechanisms and signaling pathways regulating adult SCs. We describe genetically induced-DNA damage models and highlight body-wide signals affecting cellular decisions such as survival, proliferation, and death in the presence of genomic instability. We also discuss transcriptomic changes in the DNA damage response during injury repair and propose DNA repair as key component of tissue regeneration. Additional studies using planaria will provide insights about mechanisms regulating survival and growth of cells with DNA damage during tissue renewal and regeneration.
Collapse
|
38
|
Ricci L, Srivastava M. Wound-induced cell proliferation during animal regeneration. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7:e321. [PMID: 29719123 DOI: 10.1002/wdev.321] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022]
Abstract
Many animal species are capable of replacing missing tissues that are lost upon injury or amputation through the process of regeneration. Although the extent of regeneration is variable across animals, that is, some animals can regenerate any missing cell type whereas some can only regenerate certain organs or tissues, regulated cell proliferation underlies the formation of new tissues in most systems. Notably, many species display an increase in proliferation within hours or days upon wounding. While different cell types proliferate in response to wounding in various animal taxa, comparative molecular data are beginning to point to shared wound-induced mechanisms that regulate cell division during regeneration. Here, we synthesize current insights about early molecular pathways of regeneration from diverse model and emerging systems by considering these species in their evolutionary contexts. Despite the great diversity of mechanisms underlying injury-induced cell proliferation across animals, and sometimes even in the same species, similar pathways for proliferation have been implicated in distantly related species (e.g., small diffusible molecules, signaling from apoptotic cells, growth factor signaling, mTOR and Hippo signaling, and Wnt and Bmp pathways). Studies that explicitly interrogate molecular and cellular regenerative mechanisms in understudied animal phyla will reveal the extent to which early pathways in the process of regeneration are conserved or independently evolved. This article is categorized under: Comparative Development and Evolution > Body Plan Evolution Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Comparative Development and Evolution > Model Systems.
Collapse
Affiliation(s)
- Lorenzo Ricci
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts
| | - Mansi Srivastava
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
39
|
Strand NS, Allen JM, Zayas RM. Post-translational regulation of planarian regeneration. Semin Cell Dev Biol 2018; 87:58-68. [PMID: 29705300 DOI: 10.1016/j.semcdb.2018.04.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
Most mammals cannot easily overcome degenerative disease or traumatic injuries. In contrast, an innate ability to regenerate is observed across animal phyla. Freshwater planarians are amongst the organisms that are capable of stem cell-mediated whole-body regeneration and have served as an exemplary model to study how pluripotency is maintained and regulated in vivo. Here, we review findings on the role of post-translational modifications and the genes regulating phosphorylation, ubiquitylation, and chromatin remodeling in planarian regeneration. Furthermore, we discuss how technological advances for identifying cellular targets of these processes will fill gaps in our knowledge of the signaling mechanisms that underlie regeneration in planarians, which should inform how tissue repair can be stimulated in non-regenerative model organisms and in humans.
Collapse
Affiliation(s)
- Nicholas S Strand
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - John M Allen
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Ricardo M Zayas
- Department of Biology, San Diego State University, San Diego, CA 92182, USA.
| |
Collapse
|
40
|
Neoblast-enriched zinc finger protein FIR1 triggers local proliferation during planarian regeneration. Protein Cell 2018; 10:43-59. [PMID: 29557542 PMCID: PMC6321819 DOI: 10.1007/s13238-018-0512-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 01/09/2018] [Indexed: 11/30/2022] Open
Abstract
Regeneration, relying mainly on resident adult stem cells, is widespread. However, the mechanism by which stem cells initiate proliferation during this process in vivo is unclear. Using planarian as a model, we screened 46 transcripts showing potential function in the regulation of local stem cell proliferation following 48 h regeneration. By analyzing the regeneration defects and the mitotic activity of animals under administration of RNA interference (RNAi), we identified factor for initiating regeneration 1 (Fir1) required for local proliferation. Our findings reveal that Fir1, enriched in neoblasts, promotes planarian regeneration in any tissue-missing context. Further, we demonstrate that DIS3 like 3′-5′ exoribonuclease 2 (Dis3l2) is required for Fir1 phenotype. Besides, RNAi knockdown of Fir1 causes a decrease of neoblast wound response genes following amputation. These findings suggest that Fir1 recognizes regenerative signals and promotes DIS3L2 proteins to trigger neoblast proliferation following amputation and provide a mechanism critical for stem cell response to injury.
Collapse
|
41
|
Rossi L, Salvetti A. Planarian stem cell niche, the challenge for understanding tissue regeneration. Semin Cell Dev Biol 2018. [PMID: 29534938 DOI: 10.1016/j.semcdb.2018.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Stem cell fate depends on surrounding microenvironment, the so called niche. For this reason, understanding stem cell niche is one of the most challenging target in cell biology field and need to be unraveled with in vivo studies. Planarians offer this unique opportunity, as their stem cells, the neoblasts, are abundant, highly characterized and genetically modifiable by RNA interference in alive animals. However, despite impressive advances have been done in the understanding planarian stem cells and regeneration, only a few information is available in defining signals from differentiated tissues, which affect neoblast stemness and fate. Here, we review on molecular factors that have been found activated in differentiated tissues and directly or indirectly affect neoblast behavior, and we suggest future directions for unravelling this challenge in understanding planarian stem cells.
Collapse
Affiliation(s)
- Leonardo Rossi
- Departement of Clinical and Experimental Medicine, Unit of Experimental Biology and Genetics, University of Pisa, Via Volta 4 Pisa, Italy
| | - Alessandra Salvetti
- Departement of Clinical and Experimental Medicine, Unit of Experimental Biology and Genetics, University of Pisa, Via Volta 4 Pisa, Italy.
| |
Collapse
|
42
|
Johnson K, Bateman J, DiTommaso T, Wong AY, Whited JL. Systemic cell cycle activation is induced following complex tissue injury in axolotl. Dev Biol 2018; 433:461-472. [PMID: 29111100 PMCID: PMC5750138 DOI: 10.1016/j.ydbio.2017.07.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/22/2017] [Accepted: 07/18/2017] [Indexed: 11/22/2022]
Abstract
Activation of progenitor cells is crucial to promote tissue repair following injury in adult animals. In the context of successful limb regeneration following amputation, progenitor cells residing within the stump must re-enter the cell cycle to promote regrowth of the missing limb. We demonstrate that in axolotls, amputation is sufficient to induce cell-cycle activation in both the amputated limb and the intact, uninjured contralateral limb. Activated cells were found throughout all major tissue populations of the intact contralateral limb, with internal cellular populations (bone and soft tissue) the most affected. Further, activated cells were additionally found within the heart, liver, and spinal cord, suggesting that amputation induces a common global activation signal throughout the body. Among two other injury models, limb crush and skin excisional wound, only limb crush injuries were capable of inducing cellular responses in contralateral uninjured limbs but did not achieve activation levels seen following limb loss. We found this systemic activation response to injury is independent of formation of a wound epidermis over the amputation plane, suggesting that injury-induced signals alone can promote cellular activation. In mammals, mTOR signaling has been shown to promote activation of quiescent cells following injury, and we confirmed a subset of activated contralateral cells is positive for mTOR signaling within axolotl limbs. These findings suggest that conservation of an early systemic response to injury exists between mammals and axolotls, and propose that a distinguishing feature in species capable of full regeneration is converting this initial activation into sustained and productive growth at the site of regeneration.
Collapse
Affiliation(s)
- Kimberly Johnson
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham&Women's Hospital, 60 Fenwood Rd., Boston, MA 02115, USA
| | - Joel Bateman
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham&Women's Hospital, 60 Fenwood Rd., Boston, MA 02115, USA
| | - Tia DiTommaso
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham&Women's Hospital, 60 Fenwood Rd., Boston, MA 02115, USA
| | - Alan Y Wong
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham&Women's Hospital, 60 Fenwood Rd., Boston, MA 02115, USA
| | - Jessica L Whited
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham&Women's Hospital, 60 Fenwood Rd., Boston, MA 02115, USA.
| |
Collapse
|
43
|
Strand NS, Allen JM, Ghulam M, Taylor MR, Munday RK, Carrillo M, Movsesyan A, Zayas RM. Dissecting the function of Cullin-RING ubiquitin ligase complex genes in planarian regeneration. Dev Biol 2018; 433:210-217. [PMID: 29291974 DOI: 10.1016/j.ydbio.2017.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/25/2017] [Accepted: 10/11/2017] [Indexed: 12/26/2022]
Abstract
The ubiquitin system plays a role in nearly every aspect of eukaryotic cell biology. The enzymes responsible for transferring ubiquitin onto specific substrates are the E3 ubiquitin ligases, a large and diverse family of proteins, for which biological roles and target substrates remain largely undefined. Studies using model organisms indicate that ubiquitin signaling mediates key steps in developmental processes and tissue regeneration. Here, we used the freshwater planarian, Schmidtea mediterranea, to investigate the role of Cullin-RING ubiquitin ligase (CRL) complexes in stem cell regulation during regeneration. We identified six S. mediterranea cullin genes, and used RNAi to uncover roles for homologs of Cullin-1, -3 and -4 in planarian regeneration. The cullin-1 RNAi phenotype included defects in blastema formation, organ regeneration, lesions, and lysis. To further investigate the function of cullin-1-mediated cellular processes in planarians, we examined genes encoding the adaptor protein Skp1 and F-box substrate-recognition proteins that are predicted to partner with Cullin-1. RNAi against skp1 resulted in phenotypes similar to cullin-1 RNAi, and an RNAi screen of the F-box genes identified 19 genes that recapitulated aspects of cullin-1 RNAi, including ones that in mammals are involved in stem cell regulation and cancer biology. Our data provides evidence that CRLs play discrete roles in regenerative processes and provide a platform to investigate how CRLs regulate stem cells in vivo.
Collapse
Affiliation(s)
- Nicholas S Strand
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - John M Allen
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Mahjoobah Ghulam
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Matthew R Taylor
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Roma K Munday
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Melissa Carrillo
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Artem Movsesyan
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Ricardo M Zayas
- Department of Biology, San Diego State University, San Diego, CA 92182, USA.
| |
Collapse
|
44
|
Lai AG, Kosaka N, Abnave P, Sahu S, Aboobaker AA. The abrogation of condensin function provides independent evidence for defining the self-renewing population of pluripotent stem cells. Dev Biol 2018; 433:218-226. [PMID: 28757112 PMCID: PMC5771471 DOI: 10.1016/j.ydbio.2017.07.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/18/2017] [Accepted: 07/27/2017] [Indexed: 12/18/2022]
Abstract
Heterogeneity of planarian stem cells has been categorised on the basis of single cell expression analyses and subsequent experiments to demonstrate lineage relationships. Some data suggest that despite heterogeneity in gene expression amongst cells in the cell cycle, in fact only one sub-population, known as sigma neoblasts, can self-renew. Without the tools to perform live in vivo lineage analysis, we instead took an alternative approach to provide independent evidence for defining the self-renewing stem cell population. We exploited the role of highly conserved condensin family genes to functionally assay neoblast self-renewal properties. Condensins are involved in forming properly condensed chromosomes to allow cell division to proceed during mitosis, and their abrogation inhibits mitosis and can lead to repeated endoreplication of the genome in cells that make repeated attempts to divide. We find that planarians possess only the condensin I complex, and that this is required for normal stem cell function. Abrogation of condensin function led to rapid stem cell depletion accompanied by the appearance of 'giant' cells with increased DNA content. Using previously discovered markers of heterogeneity we show that enlarged cells are always from the sigma-class of the neoblast population and we never observe evidence for endoreplication for the other neoblast subclasses. Overall, our data establish that condensins are essential for stem cell maintenance and provide independent evidence that only sigma-neoblasts are capable of multiple rounds of cell division and hence self-renewal.
Collapse
Affiliation(s)
- Alvina G Lai
- Department of Zoology, University of Oxford, Tinbergen Building, South Parks Road, Oxford OX1 3PS, United Kingdom
| | - Nobuyoshi Kosaka
- Department of Zoology, University of Oxford, Tinbergen Building, South Parks Road, Oxford OX1 3PS, United Kingdom
| | - Prasad Abnave
- Department of Zoology, University of Oxford, Tinbergen Building, South Parks Road, Oxford OX1 3PS, United Kingdom
| | - Sounak Sahu
- Department of Zoology, University of Oxford, Tinbergen Building, South Parks Road, Oxford OX1 3PS, United Kingdom
| | - A Aziz Aboobaker
- Department of Zoology, University of Oxford, Tinbergen Building, South Parks Road, Oxford OX1 3PS, United Kingdom.
| |
Collapse
|
45
|
Rink JC. Stem Cells, Patterning and Regeneration in Planarians: Self-Organization at the Organismal Scale. Methods Mol Biol 2018; 1774:57-172. [PMID: 29916155 DOI: 10.1007/978-1-4939-7802-1_2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The establishment of size and shape remains a fundamental challenge in biological research that planarian flatworms uniquely epitomize. Planarians can regenerate complete and perfectly proportioned animals from tiny and arbitrarily shaped tissue pieces; they continuously renew all organismal cell types from abundant pluripotent stem cells, yet maintain shape and anatomy in the face of constant turnover; they grow when feeding and literally degrow when starving, while scaling form and function over as much as a 40-fold range in body length or an 800-fold change in total cell numbers. This review provides a broad overview of the current understanding of the planarian stem cell system, the mechanisms that pattern the planarian body plan and how the interplay between patterning signals and cell fate choices orchestrates regeneration. What emerges is a conceptual framework for the maintenance and regeneration of the planarian body plan on basis of the interplay between pluripotent stem cells and self-organizing patterns and further, the general utility of planarians as model system for the mechanistic basis of size and shape.
Collapse
Affiliation(s)
- Jochen C Rink
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
46
|
Bryant DM, Sousounis K, Payzin-Dogru D, Bryant S, Sandoval AGW, Martinez Fernandez J, Mariano R, Oshiro R, Wong AY, Leigh ND, Johnson K, Whited JL. Identification of regenerative roadblocks via repeat deployment of limb regeneration in axolotls. NPJ Regen Med 2017; 2:30. [PMID: 29302364 PMCID: PMC5677943 DOI: 10.1038/s41536-017-0034-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 09/22/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023] Open
Abstract
Axolotl salamanders are powerful models for understanding how regeneration of complex body parts can be achieved, whereas mammals are severely limited in this ability. Factors that promote normal axolotl regeneration can be examined in mammals to determine if they exhibit altered activity in this context. Furthermore, factors prohibiting axolotl regeneration can offer key insight into the mechanisms present in regeneration-incompetent species. We sought to determine if we could experimentally compromise the axolotl's ability to regenerate limbs and, if so, discover the molecular changes that might underlie their inability to regenerate. We found that repeated limb amputation severely compromised axolotls' ability to initiate limb regeneration. Using RNA-seq, we observed that a majority of differentially expressed transcripts were hyperactivated in limbs compromised by repeated amputation, suggesting that mis-regulation of these genes antagonizes regeneration. To confirm our findings, we additionally assayed the role of amphiregulin, an EGF-like ligand, which is aberrantly upregulated in compromised animals. During normal limb regeneration, amphiregulin is expressed by the early wound epidermis, and mis-expressing this factor lead to thickened wound epithelium, delayed initiation of regeneration, and severe regenerative defects. Collectively, our results suggest that repeatedly amputated limbs may undergo a persistent wound healing response, which interferes with their ability to initiate the regenerative program. These findings have important implications for human regenerative medicine.
Collapse
Affiliation(s)
- Donald M Bryant
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Konstantinos Sousounis
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA.,The Allen Discovery Center at Tufts University, 200 Boston Ave., Suite 4600, Medford, MA 02155 USA
| | - Duygu Payzin-Dogru
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Sevara Bryant
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Aaron Gabriel W Sandoval
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Jose Martinez Fernandez
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Rachelle Mariano
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Rachel Oshiro
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Alan Y Wong
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Nicholas D Leigh
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Kimberly Johnson
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA
| | - Jessica L Whited
- Harvard Medical School, the Harvard Stem Cell Institute, and the Department of Orthopedic Surgery, Brigham and Women's Hospital, 60 Fenwood Rd., 7016D, Boston, MA 02115 USA.,The Allen Discovery Center at Tufts University, 200 Boston Ave., Suite 4600, Medford, MA 02155 USA
| |
Collapse
|
47
|
Sahu S, Dattani A, Aboobaker AA. Secrets from immortal worms: What can we learn about biological ageing from the planarian model system? Semin Cell Dev Biol 2017; 70:108-121. [PMID: 28818620 DOI: 10.1016/j.semcdb.2017.08.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 12/12/2022]
Abstract
Understanding how some animals are immortal and avoid the ageing process is important. We currently know very little about how they achieve this. Research with genetic model systems has revealed the existence of conserved genetic pathways and molecular processes that affect longevity. Most of these established model organisms have relatively short lifespans. Here we consider the use of planarians, with an immortal life-history that is able to entirely avoid the ageing process. These animals are capable of profound feats of regeneration fueled by a population of adult stem cells called neoblasts. These cells are capable of indefinite self-renewal that has underpinned the evolution of animals that reproduce only by fission, having disposed of the germline, and must therefore be somatically immortal and avoid the ageing process. How they do this is only now starting to be understood. Here we suggest that the evidence so far supports the hypothesis that the lack of ageing is an emergent property of both being highly regenerative and the evolution of highly effective mechanisms for ensuring genome stability in the neoblast stem cell population. The details of these mechanisms could prove to be very informative in understanding how the causes of ageing can be avoided, slowed or even reversed.
Collapse
Affiliation(s)
- Sounak Sahu
- Department of Zoology, South Parks Road, University of Oxford, Oxford OX1 3PS, UK
| | - Anish Dattani
- Department of Zoology, South Parks Road, University of Oxford, Oxford OX1 3PS, UK
| | - A Aziz Aboobaker
- Department of Zoology, South Parks Road, University of Oxford, Oxford OX1 3PS, UK.
| |
Collapse
|
48
|
Lin AYT, Pearson BJ. Yorkie is required to restrict the injury responses in planarians. PLoS Genet 2017; 13:e1006874. [PMID: 28686611 PMCID: PMC5515462 DOI: 10.1371/journal.pgen.1006874] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 07/18/2017] [Accepted: 06/15/2017] [Indexed: 12/30/2022] Open
Abstract
Regeneration requires the precise integration of cues that initiate proliferation, direct differentiation, and ultimately re-pattern tissues to the proper size and scale. Yet how these processes are integrated with wounding responses remains relatively unknown. The freshwater planarian, Schmidtea mediterranea, is an ideal model to study the stereotyped proliferative and transcriptional responses to injury due to its high capacity for regeneration. Here, we characterize the effector of the Hippo signalling cascade, yorkie, during planarian regeneration and its role in restricting early injury responses. In yki(RNAi) regenerating animals, wound responses are hyper-activated such that both stem cell proliferation and the transcriptional wound response program are heighted and prolonged. Using this observation, we also uncovered novel wound-induced genes by RNAseq that were de-repressed in yki(RNAi) animals compared with controls. Additionally, we show that yki(RNAi) animals have expanded epidermal and muscle cell populations, which we hypothesize are the increased sources of wound-induced genes. Finally, we show that in yki(RNAi) animals, the sensing of the size of an injury by eyes or the pharynx is not appropriate, and the brain, gut, and midline cannot remodel or scale correctly to the size of the regenerating fragment. Taken together, our results suggest that yki functions as a key molecule that can integrate multiple aspects of the injury response including proliferation, apoptosis, injury-induced transcription, and patterning. The planarian displays a remarkable ability to regenerate any tissue from mere fragments of its original size. This high capacity to regenerate is attributed to the abundant population of pluripotent adult stem cells. In response to an injury, such as an amputation, stem cells proliferate and replace the lost tissues de novo (epimorphosis), whereas existing tissue must rescale to the correct proportions in relation to the new fragment size (morphallaxis). Currently, the molecules that control either the responses to injury or the ones that mediate size and scaling are not well understood. For instance, how are the injury responses precisely activated and shut down to ensure regenerating tissues are not under- or overgrown? Here, we find that Yki, the effector of the Hippo signalling cascade, is a critical molecule that influences several injury processes during regeneration. Loss of Yki function in regenerating animals resulted in increased and temporally dysregulated expression of wound-induced genes, proliferation, and apoptosis. Genes that are injury induced were mis-expressed in yki(RNAi) animals, which also showed increases in the epidermal and muscle cell populations. Taken together, our findings suggest that the injury responses must be restricted to ensure proper regenerative outcomes of correct scale, and that Yki is a key regulator in these processes.
Collapse
Affiliation(s)
- Alexander Y. T. Lin
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Bret J. Pearson
- Hospital for Sick Children, Program in Developmental and Stem Cell Biology, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
49
|
Shettigar N, Joshi A, Dalmeida R, Gopalkrishna R, Chakravarthy A, Patnaik S, Mathew M, Palakodeti D, Gulyani A. Hierarchies in light sensing and dynamic interactions between ocular and extraocular sensory networks in a flatworm. SCIENCE ADVANCES 2017; 3:e1603025. [PMID: 28782018 PMCID: PMC5533540 DOI: 10.1126/sciadv.1603025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 06/27/2017] [Indexed: 05/23/2023]
Abstract
Light sensing has independently evolved multiple times under diverse selective pressures but has been examined only in a handful among the millions of light-responsive organisms. Unsurprisingly, mechanistic insights into how differential light processing can cause distinct behavioral outputs are limited. We show how an organism can achieve complex light processing with a simple "eye" while also having independent but mutually interacting light sensing networks. Although planarian flatworms lack wavelength-specific eye photoreceptors, a 25 nm change in light wavelength is sufficient to completely switch their phototactic behavior. Quantitative photoassays, eye-brain confocal imaging, and RNA interference/knockdown studies reveal that flatworms are able to compare small differences in the amounts of light absorbed at the eyes through a single eye opsin and convert them into binary behavioral outputs. Because planarians can fully regenerate, eye-brain injury-regeneration studies showed that this acute light intensity sensing and processing are layered on simple light detection. Unlike intact worms, partially regenerated animals with eyes can sense light but cannot sense finer gradients. Planarians also show a "reflex-like," eye-independent (extraocular/whole-body) response to low ultraviolet A light, apart from the "processive" eye-brain-mediated (ocular) response. Competition experiments between ocular and extraocular sensory systems reveal dynamic interchanging hierarchies. In intact worms, cerebral ocular response can override the reflex-like extraocular response. However, injury-regeneration again offers a time window wherein both responses coexist, but the dominance of the ocular response is reversed. Overall, we demonstrate acute light intensity-based behavioral switching and two evolutionarily distinct but interacting light sensing networks in a regenerating organism.
Collapse
Affiliation(s)
- Nishan Shettigar
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
- Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Tirumalaisamudram, Thanjavur 613401, India
| | - Asawari Joshi
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
| | - Rimple Dalmeida
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
- Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Tirumalaisamudram, Thanjavur 613401, India
| | - Rohini Gopalkrishna
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
| | - Anirudh Chakravarthy
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
| | - Siddharth Patnaik
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
| | - Manoj Mathew
- National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
| | - Dasaradhi Palakodeti
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
| | - Akash Gulyani
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences, GKVK Post, Bangalore 560065, India
| |
Collapse
|
50
|
Lai AG, Pouchkina-Stantcheva N, Di Donfrancesco A, Kildisiute G, Sahu S, Aboobaker AA. The protein subunit of telomerase displays patterns of dynamic evolution and conservation across different metazoan taxa. BMC Evol Biol 2017; 17:107. [PMID: 28441946 PMCID: PMC5405514 DOI: 10.1186/s12862-017-0949-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 04/04/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Most animals employ telomerase, which consists of a catalytic subunit known as the telomerase reverse transcriptase (TERT) and an RNA template, to maintain telomere ends. Given the importance of TERT and telomere biology in core metazoan life history traits, like ageing and the control of somatic cell proliferation, we hypothesised that TERT would have patterns of sequence and regulatory evolution reflecting the diverse life histories across the Animal Kingdom. RESULTS We performed a complete investigation of the evolutionary history of TERT across animals. We show that although TERT is almost ubiquitous across Metazoa, it has undergone substantial sequence evolution within canonical motifs. Beyond the known canonical motifs, we also identify and compare regions that are highly variable between lineages, but show conservation within phyla. Recent data have highlighted the importance of alternative splice forms of TERT in non-canonical functions and although animals may share some conserved introns, we find that the selection of exons for alternative splicing appears to be highly variable, and regulation by alternative splicing appears to be a very dynamic feature of TERT evolution. We show that even within a closely related group of triclad flatworms, where alternative splicing of TERT was previously correlated with reproductive strategy, we observe highly diverse splicing patterns. CONCLUSIONS Our work establishes that the evolutionary history and structural evolution of TERT involves previously unappreciated levels of change and the emergence of lineage specific motifs. The sequence conservation we describe within phyla suggests that these new motifs likely serve essential biological functions of TERT, which along with changes in splicing, underpin diverse functions of TERT important for animal life histories.
Collapse
Affiliation(s)
- Alvina G Lai
- Department of Zoology, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK.
| | | | | | - Gerda Kildisiute
- Department of Zoology, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK
| | - Sounak Sahu
- Department of Zoology, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK
| | - A Aziz Aboobaker
- Department of Zoology, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK.
| |
Collapse
|