1
|
Beachum AN, Salazar G, Nachbar A, Krause K, Klose H, Meyer K, Maserejian A, Ross G, Boyd H, Weigel T, Ambaye L, Miller H, Coutinho-Budd J. Glia multitask to compensate for neighboring glial cell dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611719. [PMID: 39314422 PMCID: PMC11418964 DOI: 10.1101/2024.09.06.611719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
As glia mature, they undergo glial tiling to abut one another without invading each other's boundaries. Upon the loss of the secreted neurotrophin Spätzle3 (Spz3), Drosophila cortex glia transform morphologically and lose their intricate interactions with neurons and surrounding glial subtypes. Here, we reveal that all neighboring glial cell types (astrocytes, ensheathing glia, and subperineurial glia) react by extending processes into the previous cortex glial territory to compensate for lost cortex glial function and reduce the buildup of neuronal debris. However, the loss of Spz3 alone is not sufficient for glia to cross their natural borders, as blocking CNS growth via nutrient-restriction blocks the aberrant infiltration induced by the loss of Spz3. Surprisingly, even when these neighboring glia divert their cellular resources beyond their typical borders to take on new compensatory roles, they are able to multitask to continue to preserve their own normal functions to maintain CNS homeostasis.
Collapse
Affiliation(s)
- Allison N. Beachum
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Gabriela Salazar
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Amelia Nachbar
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Kevin Krause
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Hannah Klose
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Kate Meyer
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | | | - Grace Ross
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Hannah Boyd
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Thaddeus Weigel
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Lydia Ambaye
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Hayes Miller
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Jaeda Coutinho-Budd
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
2
|
Li J, Jordana L, Mehsen H, Wang X, Archambault V. Nuclear reassembly defects after mitosis trigger apoptotic and p53-dependent safeguard mechanisms in Drosophila. PLoS Biol 2024; 22:e3002780. [PMID: 39186808 PMCID: PMC11379398 DOI: 10.1371/journal.pbio.3002780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/06/2024] [Accepted: 08/01/2024] [Indexed: 08/28/2024] Open
Abstract
In animals, mitosis involves the breakdown of the nuclear envelope and the sorting of individualized, condensed chromosomes. During mitotic exit, emerging nuclei reassemble a nuclear envelope around a single mass of interconnecting chromosomes. The molecular mechanisms of nuclear reassembly are incompletely understood. Moreover, the cellular and physiological consequences of defects in this process are largely unexplored. Here, we have characterized a mechanism essential for nuclear reassembly in Drosophila. We show that Ankle2 promotes the PP2A-dependent recruitment of BAF and Lamin at reassembling nuclei, and that failures in this mechanism result in severe nuclear defects after mitosis. We then took advantage of perturbations in this mechanism to investigate the physiological responses to nuclear reassembly defects during tissue development in vivo. Partial depletion of Ankle2, BAF, or Lamin in imaginal wing discs results in wing development defects accompanied by apoptosis. We found that blocking apoptosis strongly enhances developmental defects. Blocking p53 does not prevent apoptosis but enhances defects due to the loss of a cell cycle checkpoint. Our results suggest that apoptotic and p53-dependent responses play a crucial role in safeguarding tissue development in response to sporadic nuclear reassembly defects.
Collapse
Affiliation(s)
- Jingjing Li
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montreal, Canada
| | - Laia Jordana
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montreal, Canada
| | - Haytham Mehsen
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montreal, Canada
| | - Xinyue Wang
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
| | - Vincent Archambault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montreal, Canada
| |
Collapse
|
3
|
Zhang X, Wang Y, Zhao W, Yang S, Moussian B, Zhao Z, Zhang J, Dong W. Excess Dally-like Induces Malformation of Drosophila Legs. Cells 2024; 13:1199. [PMID: 39056781 PMCID: PMC11274743 DOI: 10.3390/cells13141199] [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: 06/05/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Glypicans are closely associated with organ development and tumorigenesis in animals. Dally-like (Dlp), a membrane-bound glypican, plays pivotal roles in various biological processes in Drosophila. In this study, we observed that an excess of Dlp led to the malformation of legs, particularly affecting the distal part. Accordingly, the leg disc was shrunken and frequently exhibited aberrant morphology. In addition, elevated Dlp levels induced ectopic cell death with no apparent cell proliferation changes. Furthermore, Dlp overexpression in the posterior compartment significantly altered Wingless (Wg) distribution. We observed a marked expansion of Wg distribution within the posterior compartment, accompanied by a corresponding decrease in the anterior compartment. It appears that excess Dlp guides Wg to diffuse to cells with higher Dlp levels. In addition, the distal-less (dll) gene, which is crucial for leg patterning, was up-regulated significantly. Notably, dachshund (dac) and homothorax (hth) expression, also essential for leg patterning and development, only appeared to be negligibly affected. Based on these findings, we speculate that excess Dlp may contribute to malformations of the distal leg region of Drosophila, possibly through its influence on Wg distribution, dll expression and induced cell death. Our research advances the understanding of Dlp function in Drosophila leg development.
Collapse
Affiliation(s)
- Xubo Zhang
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Yi Wang
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Wenting Zhao
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Shumin Yang
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Bernard Moussian
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Centre National de la Recherche Scientifique, Institut Sophia Agrobiotech, Sophia Antipolis, Université Côte d′Azur, 06108 Nice, France
| | - Zhangwu Zhao
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Jianzhen Zhang
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Wei Dong
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
4
|
Co HKC, Wu CC, Lee YC, Chen SH. Emergence of large-scale cell death through ferroptotic trigger waves. Nature 2024; 631:654-662. [PMID: 38987590 DOI: 10.1038/s41586-024-07623-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/29/2024] [Indexed: 07/12/2024]
Abstract
Large-scale cell death is commonly observed during organismal development and in human pathologies1-5. These cell death events extend over great distances to eliminate large populations of cells, raising the question of how cell death can be coordinated in space and time. One mechanism that enables long-range signal transmission is trigger waves6, but how this mechanism might be used for death events in cell populations remains unclear. Here we demonstrate that ferroptosis, an iron- and lipid-peroxidation-dependent form of cell death, can propagate across human cells over long distances (≥5 mm) at constant speeds (around 5.5 μm min-1) through trigger waves of reactive oxygen species (ROS). Chemical and genetic perturbations indicate a primary role of ROS feedback loops (Fenton reaction, NADPH oxidase signalling and glutathione synthesis) in controlling the progression of ferroptotic trigger waves. We show that introducing ferroptotic stress through suppression of cystine uptake activates these ROS feedback loops, converting cellular redox systems from being monostable to being bistable and thereby priming cell populations to become bistable media over which ROS propagate. Furthermore, we demonstrate that ferroptosis and its propagation accompany the massive, yet spatially restricted, cell death events during muscle remodelling of the embryonic avian limb, substantiating its use as a tissue-sculpting strategy during embryogenesis. Our findings highlight the role of ferroptosis in coordinating global cell death events, providing a paradigm for investigating large-scale cell death in embryonic development and human pathologies.
Collapse
Affiliation(s)
- Hannah K C Co
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
- Laboratory for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chia-Chou Wu
- Laboratory for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- National Center for Theoretical Sciences, Physics Division, Taipei, Taiwan
| | - Yi-Chen Lee
- Laboratory for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Sheng-Hong Chen
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.
- Laboratory for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
- National Center for Theoretical Sciences, Physics Division, Taipei, Taiwan.
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
5
|
Monticelli S, Sommer A, AlHajj Hassan Z, Garcia Rodriguez C, Adé K, Cattenoz P, Delaporte C, Gomez Perdiguero E, Giangrande A. Early-wave macrophages control late hematopoiesis. Dev Cell 2024; 59:1284-1301.e8. [PMID: 38569551 DOI: 10.1016/j.devcel.2024.03.013] [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: 07/06/2023] [Revised: 01/08/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
Macrophages constitute the first defense line against the non-self, but their ability to remodel their environment in organ development/homeostasis is starting to be appreciated. Early-wave macrophages (EMs), produced from hematopoietic stem cell (HSC)-independent progenitors, seed the mammalian fetal liver niche wherein HSCs expand and differentiate. The involvement of niche defects in myeloid malignancies led us to identify the cues controlling HSCs. In Drosophila, HSC-independent EMs also colonize the larva when late hematopoiesis occurs. The evolutionarily conserved immune system allowed us to investigate whether/how EMs modulate late hematopoiesis in two models. We show that loss of EMs in Drosophila and mice accelerates late hematopoiesis, which does not correlate with inflammation and does not rely on macrophage phagocytic ability. Rather, EM-derived extracellular matrix components underlie late hematopoiesis acceleration. This demonstrates a developmental role for EMs.
Collapse
Affiliation(s)
- Sara Monticelli
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, UMR, S 1258, 67400 Illkirch, France; Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, 67400 Illkirch, France
| | - Alina Sommer
- Macrophages and endothelial cells unit, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, UMR3738 CNRS, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Zeinab AlHajj Hassan
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, UMR, S 1258, 67400 Illkirch, France; Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, 67400 Illkirch, France
| | - Clarisabel Garcia Rodriguez
- Macrophages and endothelial cells unit, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, UMR3738 CNRS, 75015 Paris, France; Sorbonne Université, Collège doctoral, 75005 Paris, France
| | - Kémy Adé
- Macrophages and endothelial cells unit, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, UMR3738 CNRS, 75015 Paris, France
| | - Pierre Cattenoz
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, UMR, S 1258, 67400 Illkirch, France; Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, 67400 Illkirch, France
| | - Claude Delaporte
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, UMR, S 1258, 67400 Illkirch, France; Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, 67400 Illkirch, France
| | - Elisa Gomez Perdiguero
- Macrophages and endothelial cells unit, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, UMR3738 CNRS, 75015 Paris, France.
| | - Angela Giangrande
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, 67400 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, UMR, S 1258, 67400 Illkirch, France; Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, 67400 Illkirch, France.
| |
Collapse
|
6
|
Ahmed-de-Prado S, Estella C, Baonza A. Temporal dynamics of apoptosis-induced proliferation in pupal wing development: implications for regenerative ability. BMC Biol 2024; 22:98. [PMID: 38679694 PMCID: PMC11057159 DOI: 10.1186/s12915-024-01894-1] [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: 07/29/2023] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND The ability of animals to regenerate damaged tissue is a complex process that involves various cellular mechanisms. As animals age, they lose their regenerative abilities, making it essential to understand the underlying mechanisms that limit regenerative ability during aging. Drosophila melanogaster wing imaginal discs are epithelial structures that can regenerate after tissue injury. While significant research has focused on investigating regenerative responses during larval stages our comprehension of the regenerative potential of pupal wings and the underlying mechanisms contributing to the decline of regenerative responses remains limited. RESULTS Here, we explore the temporal dynamics during pupal development of the proliferative response triggered by the induction of cell death, a typical regenerative response. Our results indicate that the apoptosis-induced proliferative response can continue until 34 h after puparium formation (APF), beyond this point cell death alone is not sufficient to induce a regenerative response. Under normal circumstances, cell proliferation ceases around 24 h APF. Interestingly, the failure of reinitiating the cell cycle beyond this time point is not attributed to an incapacity to activate the JNK pathway. Instead, our results suggest that the function of the ecdysone-responsive transcription factor E93 is involved in limiting the apoptosis-induced proliferative response during pupal development. CONCLUSIONS Our study shows that apoptosis can prolong the proliferative period of cells in the wing during pupal development as late as 34 h APF, at least 10 h longer than during normal development. After this time point, the regenerative response is diminished, a process mediated in part by the ecdysone-responsive transcription factor E93.
Collapse
Affiliation(s)
| | - Carlos Estella
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), C/Nicolás Cabrera 1, Madrid, 28049, Spain
| | - Antonio Baonza
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), C/Nicolás Cabrera 1, Madrid, 28049, Spain.
| |
Collapse
|
7
|
Nano M, Montell DJ. Apoptotic signaling: Beyond cell death. Semin Cell Dev Biol 2024; 156:22-34. [PMID: 37988794 DOI: 10.1016/j.semcdb.2023.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/23/2023]
Abstract
Apoptosis is the best described form of regulated cell death, and was, until relatively recently, considered irreversible once particular biochemical points-of-no-return were activated. In this manuscript, we examine the mechanisms cells use to escape from a self-amplifying death signaling module. We discuss the role of feedback, dynamics, propagation, and noise in apoptotic signaling. We conclude with a revised model for the role of apoptosis in animal development, homeostasis, and disease.
Collapse
Affiliation(s)
- Maddalena Nano
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA; Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA.
| | - Denise J Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA; Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
8
|
Cumming T, Levayer R. Toward a predictive understanding of epithelial cell death. Semin Cell Dev Biol 2024; 156:44-57. [PMID: 37400292 DOI: 10.1016/j.semcdb.2023.06.008] [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: 03/30/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
Epithelial cell death is highly prevalent during development and tissue homeostasis. While we have a rather good understanding of the molecular regulators of programmed cell death, especially for apoptosis, we still fail to predict when, where, how many and which specific cells will die in a tissue. This likely relies on the much more complex picture of apoptosis regulation in a tissular and epithelial context, which entails cell autonomous but also non-cell autonomous factors, diverse feedback and multiple layers of regulation of the commitment to apoptosis. In this review, we illustrate this complexity of epithelial apoptosis regulation by describing these different layers of control, all demonstrating that local cell death probability is a complex emerging feature. We first focus on non-cell autonomous factors that can locally modulate the rate of cell death, including cell competition, mechanical input and geometry as well as systemic effects. We then describe the multiple feedback mechanisms generated by cell death itself. We also outline the multiple layers of regulation of epithelial cell death, including the coordination of extrusion and regulation occurring downstream of effector caspases. Eventually, we propose a roadmap to reach a more predictive understanding of cell death regulation in an epithelial context.
Collapse
Affiliation(s)
- Tom Cumming
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France; Sorbonne Université, Collège Doctoral, F75005 Paris, France
| | - Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
9
|
Sun G. Death and survival from executioner caspase activation. Semin Cell Dev Biol 2024; 156:66-73. [PMID: 37468421 DOI: 10.1016/j.semcdb.2023.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Executioner caspases are evolutionarily conserved regulators of cell death under apoptotic stress. Activated executioner caspases drive apoptotic cell death through cleavage of diverse protein substrates or pyroptotic cell death in the presence of gasdermin E. On the other hand, activation of executioner caspases can also trigger pro-survival and pro-proliferation signals. In recent years, a growing body of studies have demonstrated that cells can survive from executioner caspase activation in response to stress and that the survivors undergo molecular and phenotypic alterations. This review focuses on death and survival from executioner caspase activation, summarizing the role of executioner caspases in apoptotic and pyroptotic cell death and discussing the potential mechanism and consequences of survival from stress-induced executioner caspase activation.
Collapse
Affiliation(s)
- Gongping Sun
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
10
|
Brown J, Su TT. E2F1, DIAP1, and the presence of a homologous chromosome promote while JNK inhibits radiation-induced loss of heterozygosity in Drosophila melanogaster. Genetics 2024; 226:iyad192. [PMID: 37874851 PMCID: PMC10763536 DOI: 10.1093/genetics/iyad192] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 08/25/2023] [Accepted: 10/04/2023] [Indexed: 10/26/2023] Open
Abstract
Loss of heterozygosity (LOH) can occur when a heterozygous mutant cell loses the remaining wild-type allele to become a homozygous mutant. LOH can have physiological consequences if, for example, the affected gene encodes a tumor suppressor. We used fluorescent reporters to study the mechanisms of LOH induction by X-rays, a type of ionizing radiation (IR), in Drosophila melanogaster larval wing discs. IR is used to treat more than half of patients with cancer, so understanding its effects is of biomedical relevance. Quantitative analysis of IR-induced LOH at different positions between the telomere and the centromere on the X chromosome showed a strong sex dependence and the need for a recombination-proficient homologous chromosome, whereas, paradoxically, position along the chromosome made little difference in LOH incidence. We propose that published data documenting high recombination frequency within centromeric heterochromatin on the X chromosome can explain these data. Using a focused screen, we identified E2F1 as a key promotor of LOH and further testing suggests a mechanism involving its role in cell-cycle regulation. We leveraged the loss of a transcriptional repressor through LOH to express transgenes specifically in cells that have already acquired LOH. This approach identified JNK signaling and apoptosis as key determinants of LOH maintenance. These studies reveal previously unknown mechanisms for the generation and elimination of cells with chromosome aberrations after exposure to IR.
Collapse
Affiliation(s)
- Jeremy Brown
- Department of Molecular, Cellular and Developmental Biology, 347 UCB, University of Colorado, Boulder, CO 80309-0347, USA
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, 13001 E. 17th Pl., Aurora, CO 80045, USA
| | - Tin Tin Su
- Department of Molecular, Cellular and Developmental Biology, 347 UCB, University of Colorado, Boulder, CO 80309-0347, USA
- University of Colorado Cancer Center, Anschutz Medical Campus, 13001 E. 17th Pl., Aurora, CO 80045, USA
| |
Collapse
|
11
|
Boone M, Zappa F. Signaling plasticity in the integrated stress response. Front Cell Dev Biol 2023; 11:1271141. [PMID: 38143923 PMCID: PMC10740175 DOI: 10.3389/fcell.2023.1271141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/29/2023] [Indexed: 12/26/2023] Open
Abstract
The Integrated Stress Response (ISR) is an essential homeostatic signaling network that controls the cell's biosynthetic capacity. Four ISR sensor kinases detect multiple stressors and relay this information to downstream effectors by phosphorylating a common node: the alpha subunit of the eukaryotic initiation factor eIF2. As a result, general protein synthesis is repressed while select transcripts are preferentially translated, thus remodeling the proteome and transcriptome. Mounting evidence supports a view of the ISR as a dynamic signaling network with multiple modulators and feedback regulatory features that vary across cell and tissue types. Here, we discuss updated views on ISR sensor kinase mechanisms, how the subcellular localization of ISR components impacts signaling, and highlight ISR signaling differences across cells and tissues. Finally, we consider crosstalk between the ISR and other signaling pathways as a determinant of cell health.
Collapse
|
12
|
Yusupova M, Ankawa R, Yosefzon Y, Meiri D, Bachelet I, Fuchs Y. Apoptotic dysregulation mediates stem cell competition and tissue regeneration. Nat Commun 2023; 14:7547. [PMID: 37985759 PMCID: PMC10662150 DOI: 10.1038/s41467-023-41684-x] [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: 08/25/2022] [Accepted: 09/14/2023] [Indexed: 11/22/2023] Open
Abstract
Since adult stem cells are responsible for replenishing tissues throughout life, it is vital to understand how failure to undergo apoptosis can dictate stem cell behavior both intrinsically and non-autonomously. Here, we report that depletion of pro-apoptotic Bax protein bestows hair follicle stem cells with the capacity to eliminate viable neighboring cells by sequestration of TNFα in their membrane. This in turn induces apoptosis in "loser" cells in a contact-dependent manner. Examining the underlying mechanism, we find that Bax loss-of-function competitive phenotype is mediated by the intrinsic activation of NFκB. Notably, winner stem cells differentially respond to TNFα, owing to their elevated expression of TNFR2. Finally, we report that in vivo depletion of Bax results in an increased stem cell pool, accelerating wound-repair and de novo hair follicle regeneration. Collectively, we establish a mechanism of mammalian cell competition, which can have broad therapeutic implications for tissue regeneration and tumorigenesis.
Collapse
Affiliation(s)
- Marianna Yusupova
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Roi Ankawa
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
- Augmanity, Rehovot, Israel
| | - Yahav Yosefzon
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - David Meiri
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Yaron Fuchs
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
- Augmanity, Rehovot, Israel.
| |
Collapse
|
13
|
Martínez-Abarca Millán A, Martín-Bermudo MD. Integrins Can Act as Suppressors of Ras-Mediated Oncogenesis in the Drosophila Wing Disc Epithelium. Cancers (Basel) 2023; 15:5432. [PMID: 38001693 PMCID: PMC10670217 DOI: 10.3390/cancers15225432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is the second leading cause of death worldwide. Key to cancer initiation and progression is the crosstalk between cancer cells and their microenvironment. The extracellular matrix (ECM) is a major component of the tumour microenvironment and integrins, main cell-ECM adhesion receptors, are involved in every step of cancer progression. However, accumulating evidence has shown that integrins can act as tumour promoters but also as tumour suppressor factors, revealing that the biological roles of integrins in cancer are complex. This incites a better understating of integrin function in cancer progression. To achieve this goal, simple model organisms, such as Drosophila, offer great potential to unravel underlying conceptual principles. Here, we find that in the Drosophila wing disc epithelium the βPS integrins act as suppressors of tumours induced by a gain of function of the oncogenic form of Ras, RasV12. We show that βPS integrin depletion enhances the growth, delamination and invasive behaviour of RasV12 tumour cells, as well as their ability to affect the tumour microenvironment. These results strongly suggest that integrin function as tumour suppressors might be evolutionarily conserved. Drosophila can be used to understand the complex tumour modulating activities conferred by integrins, thus facilitating drug development.
Collapse
Affiliation(s)
| | - María D. Martín-Bermudo
- Centro Andaluz de Biología del Desarrollo, CSIC, Universidad Pablo de Olavide, 41013 Sevilla, Spain
| |
Collapse
|
14
|
Gregory CD. Hijacking homeostasis: Regulation of the tumor microenvironment by apoptosis. Immunol Rev 2023; 319:100-127. [PMID: 37553811 PMCID: PMC10952466 DOI: 10.1111/imr.13259] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023]
Abstract
Cancers are genetically driven, rogue tissues which generate dysfunctional, obdurate organs by hijacking normal, homeostatic programs. Apoptosis is an evolutionarily conserved regulated cell death program and a profoundly important homeostatic mechanism that is common (alongside tumor cell proliferation) in actively growing cancers, as well as in tumors responding to cytotoxic anti-cancer therapies. Although well known for its cell-autonomous tumor-suppressive qualities, apoptosis harbors pro-oncogenic properties which are deployed through non-cell-autonomous mechanisms and which generally remain poorly defined. Here, the roles of apoptosis in tumor biology are reviewed, with particular focus on the secreted and fragmentation products of apoptotic tumor cells and their effects on tumor-associated macrophages, key supportive cells in the aberrant homeostasis of the tumor microenvironment. Historical aspects of cell loss in tumor growth kinetics are considered and the impact (and potential impact) on tumor growth of apoptotic-cell clearance (efferocytosis) as well as released soluble and extracellular vesicle-associated factors are discussed from the perspectives of inflammation, tissue repair, and regeneration programs. An "apoptosis-centric" view is proposed in which dying tumor cells provide an important platform for intricate intercellular communication networks in growing cancers. The perspective has implications for future research and for improving cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Christopher D. Gregory
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarterEdinburghUK
| |
Collapse
|
15
|
Nagai H, Nagai LAE, Tasaki S, Nakato R, Umetsu D, Kuranaga E, Miura M, Nakajima Y. Nutrient-driven dedifferentiation of enteroendocrine cells promotes adaptive intestinal growth in Drosophila. Dev Cell 2023; 58:1764-1781.e10. [PMID: 37689060 DOI: 10.1016/j.devcel.2023.08.022] [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: 07/22/2022] [Revised: 05/05/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Post-developmental organ resizing improves organismal fitness under constantly changing nutrient environments. Although stem cell abundance is a fundamental determinant of adaptive resizing, our understanding of its underlying mechanisms remains primarily limited to the regulation of stem cell division. Here, we demonstrate that nutrient fluctuation induces dedifferentiation in the Drosophila adult midgut to drive adaptive intestinal growth. From lineage tracing and single-cell RNA sequencing, we identify a subpopulation of enteroendocrine (EE) cells that convert into functional intestinal stem cells (ISCs) in response to dietary glucose and amino acids by activating the JAK-STAT pathway. Genetic ablation of EE-derived ISCs severely impairs ISC expansion and midgut growth despite the retention of resident ISCs, and in silico modeling further indicates that EE dedifferentiation enables an efficient increase in the midgut cell number while maintaining epithelial cell composition. Our findings identify a physiologically induced dedifferentiation that ensures ISC expansion during adaptive organ growth in concert with nutrient conditions.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| | | | - Sohei Tasaki
- Graduate School of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Ryuichiro Nakato
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Erina Kuranaga
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuichiro Nakajima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| |
Collapse
|
16
|
Villars A, Letort G, Valon L, Levayer R. DeXtrusion: automatic recognition of epithelial cell extrusion through machine learning in vivo. Development 2023; 150:dev201747. [PMID: 37283069 PMCID: PMC10323232 DOI: 10.1242/dev.201747] [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: 03/03/2023] [Accepted: 05/28/2023] [Indexed: 06/08/2023]
Abstract
Accurately counting and localising cellular events from movies is an important bottleneck of high-content tissue/embryo live imaging. Here, we propose a new methodology based on deep learning that allows automatic detection of cellular events and their precise xyt localisation on live fluorescent imaging movies without segmentation. We focused on the detection of cell extrusion, the expulsion of dying cells from the epithelial layer, and devised DeXtrusion: a pipeline based on recurrent neural networks for automatic detection of cell extrusion/cell death events in large movies of epithelia marked with cell contour. The pipeline, initially trained on movies of the Drosophila pupal notum marked with fluorescent E-cadherin, is easily trainable, provides fast and accurate extrusion predictions in a large range of imaging conditions, and can also detect other cellular events, such as cell division or cell differentiation. It also performs well on other epithelial tissues with reasonable re-training. Our methodology could easily be applied for other cellular events detected by live fluorescent microscopy and could help to democratise the use of deep learning for automatic event detections in developing tissues.
Collapse
Affiliation(s)
- Alexis Villars
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France
| | - Gaëlle Letort
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France
| | - Léo Valon
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France
| | - Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
17
|
Brown J, Su TT. E2F1 promotes, JNK and DIAP1 inhibit, and chromosomal position has little effect on radiation-induced Loss of Heterozygosity in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.540084. [PMID: 37214983 PMCID: PMC10197609 DOI: 10.1101/2023.05.09.540084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Loss of Heterozygosity (LOH) can occur when a heterozygous mutant cell loses the remaining wild type allele to become a homozygous mutant. LOH can have physiological consequences if, for example, the affected gene encodes a tumor suppressor. We used two fluorescent reporters to study mechanisms of LOH induction by X-rays, a type of ionizing radiation (IR), in Drosophila larval wing discs. IR is used to treat more than half of cancer patients, so understanding its effects is of biomedical relevance. We report that IR-induced LOH does not correlate with the chromosomal position of the LOH locus, unlike previously shown for spontaneously occurring LOH. Like spontaneous LOH, however, IR-induced LOH of X-linked loci shows a sex-dependence, occurring predominately in females. A focused genetic screen identified E2F1 as a key promotor of LOH and further testing suggests a mechanism involving its role in cell cycle regulation rather than apoptosis. We combined the QF/QS LOH reporter with QUAS-transgenes to manipulate gene function after LOH induction. This approach identified JNK signaling and apoptosis as key determinants of LOH maintenance. These studies reveal previously unknown mechanisms for generation and maintenance of cells with chromosome aberrations after exposure to IR.
Collapse
|
18
|
Yusupova M, Fuchs Y. To not love thy neighbor: mechanisms of cell competition in stem cells and beyond. Cell Death Differ 2023; 30:979-991. [PMID: 36813919 PMCID: PMC10070350 DOI: 10.1038/s41418-023-01114-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 02/24/2023] Open
Abstract
Cell competition describes the process in which cells of greater fitness are capable of sensing and instructing elimination of lesser fit mutant cells. Since its discovery in Drosophila, cell competition has been established as a critical regulator of organismal development, homeostasis, and disease progression. It is therefore unsurprising that stem cells (SCs), which are central to these processes, harness cell competition to remove aberrant cells and preserve tissue integrity. Here, we describe pioneering studies of cell competition across a variety of cellular contexts and organisms, with the ultimate goal of better understanding competition in mammalian SCs. Furthermore, we explore the modes through which SC competition takes place and how this facilitates normal cellular function or contributes to pathological states. Finally, we discuss how understanding of this critical phenomenon will enable targeting of SC-driven processes, including regeneration and tumor progression.
Collapse
Affiliation(s)
- Marianna Yusupova
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel.
- Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Haifa, Israel.
- Augmanity, Rehovot, Israel.
| |
Collapse
|
19
|
Duran C, Barcenas M, Wang Q. Modeling of ionizing radiation induced hair follicle regenerative dynamics. J Theor Biol 2022; 555:111283. [PMID: 36181867 PMCID: PMC10151310 DOI: 10.1016/j.jtbi.2022.111283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/27/2022] [Accepted: 09/16/2022] [Indexed: 01/14/2023]
Abstract
Hair follicles (HFs) are stem-cell-rich mammalian mini-organs that can undergo cyclic regenerations over the life span of the organism. The cycle of a HF consists of three consecutive phases: anagen-the active proliferation phase, catagen-the degeneration phase, and telogen-the resting phase. While HFs undergo irreversible degeneration during catagen, recent experimental research on mice shows that when anagen HFs are subject to ionizing radiation (IR), they undergo a transient degeneration, followed by a nearly full regeneration that makes the HFs return to homeostatic state. The mechanisms underlying these IR-induced HF regenerative dynamics and the catagen degenerative dynamics, remain unknown. In this work, we develop an ODE type cell differentiation population model to study the control mechanisms of HF regeneration. The model is built based on current theoretical knowledge in biology and mathematically formulated using feedback mechanisms. Model parameters are calibrated to IR experimental data, and we then provide modeling results with both deterministic ODE simulations and corresponding stochastic simulations. We perform stability and bifurcation analyses on the ODE model, which reveal that for anagen HFs, a low spontaneous apoptosis rate secures the stability of the HF homeostatic steady state, allowing the HF to regenerate even when subject to strong IR. On the other hand, the irreversible degeneration during catagen results from both strong spontaneous apoptosis rate and strong apoptosis feedback. Lastly, we perform sensitivity analysis to identify key parameters in the model to validate these hypotheses.
Collapse
Affiliation(s)
- Cecilia Duran
- Department of Mathematics, University of California, Riverside, CA, USA; Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, USA
| | - Manuel Barcenas
- Department of Mathematics, University of California, Riverside, CA, USA
| | - Qixuan Wang
- Department of Mathematics, University of California, Riverside, CA, USA; Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, USA.
| |
Collapse
|
20
|
Coursier D, Coulette D, Leman H, Grenier E, Ichim G. Live-cell imaging and mathematical analysis of the “community effect” in apoptosis. Apoptosis 2022; 28:326-334. [PMID: 36346539 DOI: 10.1007/s10495-022-01783-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2022] [Indexed: 11/11/2022]
Abstract
As a cellular intrinsic mechanism leading to cellular demise, apoptosis was thoroughly characterized from a mechanistic perspective. Nowadays there is an increasing interest in describing the non-cell autonomous or community effects of apoptosis, especially in the context of resistance to cancer treatments. Transitioning from cell-centered to cell population-relevant mechanisms adds a layer of complexity for imaging and analyzing an enormous number of apoptotic events. In addition, the community effect between apoptotic and living cells is difficult to be taken into account for complex analysis. We describe here a robust and easy to implement method to analyze the interactions between cancer cells, while under apoptotic pressure. Using this approach we showed as proof-of-concept that apoptosis is insensitive to cellular density, while the proximity to apoptotic cells increases the probability of a given cell to undergo apoptosis.
Collapse
Affiliation(s)
- Diane Coursier
- Cancer Research Center of Lyon (CRCL) INSERM 1052, CNRS 5286, Lyon, France
- Cancer Cell Death Laboratory, Part of LabEx DEVweCAN, Université de Lyon, Lyon, France
| | - David Coulette
- ENS-Lyon, UMR CNRS 5669 'UMPA' and INRIA Lyon, Project NUMED, Lyon, 69364, France
| | - Hélène Leman
- ENS-Lyon, UMR CNRS 5669 'UMPA' and INRIA Lyon, Project NUMED, Lyon, 69364, France
| | - Emmanuel Grenier
- ENS-Lyon, UMR CNRS 5669 'UMPA' and INRIA Lyon, Project NUMED, Lyon, 69364, France
| | - Gabriel Ichim
- Cancer Research Center of Lyon (CRCL) INSERM 1052, CNRS 5286, Lyon, France.
- Cancer Cell Death Laboratory, Part of LabEx DEVweCAN, Université de Lyon, Lyon, France.
| |
Collapse
|
21
|
Riley JS, Bock FJ. Voices from beyond the grave: The impact of apoptosis on the microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119341. [PMID: 35987283 DOI: 10.1016/j.bbamcr.2022.119341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 08/09/2022] [Indexed: 06/15/2023]
Abstract
Programmed cell death, in particular apoptosis, has vital functions in every healthy organism. In a highly regulated manner cells which are no longer needed or are harmful to the organism undergo suicide. More than just the mere elimination of a cell, apoptosis is increasingly being recognized performing important roles in cellular communication with the microenvironment. These interactions with surrounding cells can have various, and sometimes competing outcomes. Apoptotic cells can promote survival, proliferation and inflammation, but depending on the context also prevent survival and inflammation. In this review, we will summarize the emerging literature on how dying cells can transfer information to their neighbours, and which outcomes this communication has for the whole tissue.
Collapse
Affiliation(s)
- Joel S Riley
- Institute of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| | - Florian J Bock
- Department of Radiotherapy (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
22
|
Worley MI, Hariharan IK. Imaginal Disc Regeneration: Something Old, Something New. Cold Spring Harb Perspect Biol 2022; 14:a040733. [PMID: 34872971 PMCID: PMC9620854 DOI: 10.1101/cshperspect.a040733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Imaginal discs are simple epithelial sacs found in Drosophila larvae, which generate adult structures including wings and legs. The first studies of imaginal disc regeneration involved technically challenging transplantation experiments. Yet despite the difficulty, many aspects of regeneration including wound healing, blastema formation, and the repatterning of regenerated tissue were characterized. An important discovery was the phenomenon of transdetermination, where a small group of cells in regenerating tissue collectively switch fate ("collective cell reprogramming"). The development of genetic tissue-ablation systems over the last 12 years has energized this field, by making experiments less technically challenging, more reproducible, and by incorporating additional genetic analysis. Recent progress includes defining mechanistic links between early responses to wounding and the signaling pathways that drive proliferation, uncovering a role for localized silencing of damage-responsive enhancers to limit regenerative capacity as tissues mature, and identifying genes that maintain cellular plasticity within acceptable limits during regeneration.
Collapse
Affiliation(s)
- Melanie I Worley
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200, USA
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200, USA
| |
Collapse
|
23
|
Evans CJ, Bieser KL, Acevedo-Vasquez KS, Augustine EJ, Bowen S, Casarez VA, Feliciano VI, Glazier A, Guinan HR, Hallman R, Haugan E, Hehr LA, Hunnicutt SN, Leifer I, Mauger M, Mauger M, Melendez NY, Milshteyn L, Moore E, Nguyen SA, Phanphouvong SC, Pinal DM, Pope HM, Salinas MBM, Shellin M, Small I, Yeoh NC, Yokomizo AM, Kagey JD. The I.3.2 developmental mutant has a single nucleotide deletion in the gene centromere identifier. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000653. [PMID: 36389120 PMCID: PMC9644223 DOI: 10.17912/micropub.biology.000653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/28/2022] [Accepted: 10/16/2022] [Indexed: 06/16/2023]
Abstract
The mutation I.3.2 was previously identified in a FLP/FRT screen of chromosome 2R for conditional growth regulators. Here we report the phenotypic characterization and genetic mapping of I.3.2 by undergraduate students participating in Fly-CURE, a pedagogical program that teaches the science of genetics through a classroom research experience. We find that creation of I.3.2 cell clones in the developing eye-antennal imaginal disc causes a headless adult phenotype, suggestive of both autonomous and non-autonomous effects on cell growth or viability. We also identify the I.3.2 mutation as a loss-of-function allele of the gene centromere identifier ( cid ), which encodes centromere-specific histone H3 variant critical for chromosomal segregation.
Collapse
Affiliation(s)
- Cory J. Evans
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Kayla L. Bieser
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | | | - Emyli J. Augustine
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Skyler Bowen
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | | | - Vanessa I. Feliciano
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Ashley Glazier
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Haley R. Guinan
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Randy Hallman
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Elizabeth Haugan
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Lauren A. Hehr
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Shawna N. Hunnicutt
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Isabella Leifer
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Meaghan Mauger
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Morgan Mauger
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Norma Y. Melendez
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Larry Milshteyn
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Eric Moore
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Sarah A. Nguyen
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | | | - David M. Pinal
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | - Hailee M. Pope
- Department of Physical and Life Sciences, Nevada State College, Henderson, NV, USA
| | | | - Matthew Shellin
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Ivana Small
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | - Neelufar C. Yeoh
- Department of Biology, Loyola Marymount University, Los Angeles, CA, USA
| | | | - Jacob D. Kagey
- Biology Department, University of Detroit Mercy, Detroit, MI, USA
| |
Collapse
|
24
|
Vinestock RC, Felsenthal N, Assaraf E, Katz E, Rubin S, Heinemann-Yerushalmi L, Krief S, Dezorella N, Levin-Zaidman S, Tsoory M, Thomopoulos S, Zelzer E. Neonatal Enthesis Healing Involves Noninflammatory Acellular Scar Formation through Extracellular Matrix Secretion by Resident Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1122-1135. [PMID: 35659946 PMCID: PMC9379688 DOI: 10.1016/j.ajpath.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/19/2022] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Wound healing typically recruits the immune and vascular systems to restore tissue structure and function. However, injuries to the enthesis, a hypocellular and avascular tissue, often result in fibrotic scar formation and loss of mechanical properties, severely affecting musculoskeletal function and life quality. This raises questions about the healing capabilities of the enthesis. Herein, this study established an injury model to the Achilles entheses of neonatal mice to study the effectiveness of early-age enthesis healing. Histology and immunohistochemistry analyses revealed an atypical process that did not involve inflammation or angiogenesis. Instead, healing was mediated by secretion of collagen types I and II by resident cells, which formed a permanent hypocellular and avascular scar. Transmission electron microscopy showed that the cellular response to injury, including endoplasmic reticulum stress, autophagy, and cell death, varied between the tendon and cartilage ends of the enthesis. Single-molecule in situ hybridization, immunostaining, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays verified these differences. Finally, gait analysis showed that these processes effectively restored function of the injured leg. These findings reveal a novel healing mechanism in neonatal entheses, whereby local extracellular matrix secretion by resident cells forms an acellular extracellular matrix deposit without inflammation, allowing gait restoration. These insights into the healing mechanism of a complex transitional tissue may lead to new therapeutic strategies for adult enthesis injuries.
Collapse
Affiliation(s)
- Ron C Vinestock
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Assaraf
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Katz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nili Dezorella
- Department of Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Smadar Levin-Zaidman
- Department of Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, New York; Department of Biomedical Engineering, Columbia University, New York, New York
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
25
|
A probabilistic Boolean model on hair follicle cell fate regulation by TGF-β. Biophys J 2022; 121:2638-2652. [PMID: 35714600 DOI: 10.1016/j.bpj.2022.05.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
Hair follicles (HFs) are mini skin organs that undergo cyclic growth. Various signals regulate HF cell fate decisions jointly. Recent experimental results suggest that transforming growth factor beta (TGF-β) exhibits a dual role in HF cell fate regulation that can be either anti- or pro-apoptosis. To understand the underlying mechanisms of HF cell fate control, we develop a novel probabilistic Boolean network (pBN) model on the HF epithelial cell gene regulation dynamics. First, the model is derived from literature, then refined using single-cell RNA sequencing data. Using the model, we both explore the mechanisms underlying HF cell fate decisions and make predictions that could potentially guide future experiments: 1) we propose that a threshold-like switch in the TGF-β strength may necessitate the dual roles of TGF-β in either activating apoptosis or cell proliferation, in cooperation with Bmp and tumor necrosis factor (TNF) and at different stages of a follicle growth cycle; 2) our model shows concordance with the high-activator-low-inhibitor theory of anagen initiation; 3) we predict that TNF may be more effective in catagen initiation than TGF-β, and they may cooperate in a two-step fashion; 4) finally, predictions of gene knockout and overexpression reveal the roles in HF cell fate regulations of each gene. Attractor and motif analysis from the associated Boolean networks reveal the relations between the topological structure of the gene regulation network and the cell fate regulation mechanism. A discrete spatial model equipped with the pBN illustrates how TGF-β and TNF cooperate in initiating and driving the apoptosis wave during catagen.
Collapse
|
26
|
Avalos PN, Forsthoefel DJ. An Emerging Frontier in Intercellular Communication: Extracellular Vesicles in Regeneration. Front Cell Dev Biol 2022; 10:849905. [PMID: 35646926 PMCID: PMC9130466 DOI: 10.3389/fcell.2022.849905] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Regeneration requires cellular proliferation, differentiation, and other processes that are regulated by secreted cues originating from cells in the local environment. Recent studies suggest that signaling by extracellular vesicles (EVs), another mode of paracrine communication, may also play a significant role in coordinating cellular behaviors during regeneration. EVs are nanoparticles composed of a lipid bilayer enclosing proteins, nucleic acids, lipids, and other metabolites, and are secreted by most cell types. Upon EV uptake by target cells, EV cargo can influence diverse cellular behaviors during regeneration, including cell survival, immune responses, extracellular matrix remodeling, proliferation, migration, and differentiation. In this review, we briefly introduce the history of EV research and EV biogenesis. Then, we review current understanding of how EVs regulate cellular behaviors during regeneration derived from numerous studies of stem cell-derived EVs in mammalian injury models. Finally, we discuss the potential of other established and emerging research organisms to expand our mechanistic knowledge of basic EV biology, how injury modulates EV biogenesis, cellular sources of EVs in vivo, and the roles of EVs in organisms with greater regenerative capacity.
Collapse
Affiliation(s)
- Priscilla N. Avalos
- Department of Cell Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - David J. Forsthoefel
- Department of Cell Biology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| |
Collapse
|
27
|
Xu DC, Wang L, Yamada KM, Baena-Lopez LA. Non-apoptotic activation of Drosophila caspase-2/9 modulates JNK signaling, the tumor microenvironment, and growth of wound-like tumors. Cell Rep 2022; 39:110718. [PMID: 35443185 PMCID: PMC9082238 DOI: 10.1016/j.celrep.2022.110718] [Citation(s) in RCA: 4] [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: 08/04/2020] [Revised: 02/15/2022] [Accepted: 03/31/2022] [Indexed: 11/23/2022] Open
Abstract
Resistance to apoptosis due to caspase deregulation is considered one of the main hallmarks of cancer. However, the discovery of novel non-apoptotic caspase functions has revealed unknown intricacies about the interplay between these enzymes and tumor progression. To investigate this biological problem, we capitalized on a Drosophila tumor model with human relevance based on the simultaneous overactivation of the EGFR and the JAK/STAT signaling pathways. Our data indicate that widespread non-apoptotic activation of initiator caspases limits JNK signaling and facilitates cell fate commitment in these tumors, thus preventing the overgrowth and exacerbation of malignant features of transformed cells. Intriguingly, caspase activity also reduces the presence of macrophage-like cells with tumor-promoting properties in the tumor microenvironment. These findings assign tumor-suppressing activities to caspases independent of apoptosis, while providing molecular details to better understand the contribution of these enzymes to tumor progression.
Collapse
Affiliation(s)
- Derek Cui Xu
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, USA; Sir William Dunn School of Pathology, University of Oxford, Oxford, Oxfordshire OX1 3RE, UK
| | - Li Wang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, Oxfordshire OX1 3RE, UK
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, USA.
| | | |
Collapse
|
28
|
Roellig D, Theis S, Proag A, Allio G, Bénazéraf B, Gros J, Suzanne M. Force-generating apoptotic cells orchestrate avian neural tube bending. Dev Cell 2022; 57:707-718.e6. [PMID: 35303434 PMCID: PMC8967407 DOI: 10.1016/j.devcel.2022.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 11/15/2021] [Accepted: 02/22/2022] [Indexed: 12/16/2022]
Abstract
Apoptosis plays an important role in morphogenesis, and the notion that apoptotic cells can impact their surroundings came to light recently. However, how this applies to vertebrate morphogenesis remains unknown. Here, we use the formation of the neural tube to determine how apoptosis contributes to morphogenesis in vertebrates. Neural tube closure defects have been reported when apoptosis is impaired in vertebrates, although the cellular mechanisms involved are unknown. Using avian embryos, we found that apoptotic cells generate an apico-basal force before being extruded from the neuro-epithelium. This force, which relies on a contractile actomyosin cable that extends along the apico-basal axis of the cell, drives nuclear fragmentation and influences the neighboring tissue. Together with the morphological defects observed when apoptosis is prevented, these data strongly suggest that the neuroepithelium keeps track of the mechanical impact of apoptotic cells and that the apoptotic forces, cumulatively, contribute actively to neural tube bending. Apoptotic cells are force-generating cells in the avian neural tube Apoptotic force drives the upward movement of the nucleus and nuclear fragmentation Apoptotic cells cumulatively impact the neighboring tissue Apoptotic force mechanical impact participates in progressive bending of the neural tube
Collapse
Affiliation(s)
- Daniela Roellig
- Centre de Biologie Intégrative, CNRS/UMR 5088, Université Toulouse III, Toulouse, France
| | - Sophie Theis
- Centre de Biologie Intégrative, CNRS/UMR 5088, Université Toulouse III, Toulouse, France; Morphogénie Logiciels, 32110 St Martin d'Armagnac, France
| | - Amsha Proag
- Centre de Biologie Intégrative, CNRS/UMR 5088, Université Toulouse III, Toulouse, France
| | - Guillaume Allio
- Centre de Biologie Intégrative, CNRS/UMR 5088, Université Toulouse III, Toulouse, France
| | - Bertrand Bénazéraf
- Centre de Biologie Intégrative, CNRS/UMR 5088, Université Toulouse III, Toulouse, France
| | - Jérôme Gros
- Institut Pasteur, CNRS/UMR 3738, Paris, France
| | - Magali Suzanne
- Centre de Biologie Intégrative, CNRS/UMR 5088, Université Toulouse III, Toulouse, France.
| |
Collapse
|
29
|
Liu KE, Raymond MH, Ravichandran KS, Kucenas S. Clearing Your Mind: Mechanisms of Debris Clearance After Cell Death During Neural Development. Annu Rev Neurosci 2022; 45:177-198. [PMID: 35226828 PMCID: PMC10157384 DOI: 10.1146/annurev-neuro-110920-022431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurodevelopment and efferocytosis have fascinated scientists for decades. How an organism builds a nervous system that is precisely tuned for efficient behaviors and survival and how it simultaneously manages constant somatic cell turnover are complex questions that have resulted in distinct fields of study. Although neurodevelopment requires the overproduction of cells that are subsequently pruned back, very few studies marry these fields to elucidate the cellular and molecular mechanisms that drive nervous system development through the lens of cell clearance. In this review, we discuss these fields to highlight exciting areas of future synergy. We first review neurodevelopment from the perspective of overproduction and subsequent refinement and then discuss who clears this developmental debris and the mechanisms that control these events. We then end with how a more deliberate merger of neurodevelopment and efferocytosis could reframe our understanding of homeostasis and disease and discuss areas of future study. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kendra E Liu
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Michael H Raymond
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Center for Clearance, University of Virginia, Charlottesville, Virginia, USA
| | - Kodi S Ravichandran
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Center for Clearance, University of Virginia, Charlottesville, Virginia, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA.,VIB-UGent Center for Inflammation Research and the Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sarah Kucenas
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA.,Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
30
|
Morana O, Wood W, Gregory CD. The Apoptosis Paradox in Cancer. Int J Mol Sci 2022; 23:ijms23031328. [PMID: 35163253 PMCID: PMC8836235 DOI: 10.3390/ijms23031328] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer growth represents a dysregulated imbalance between cell gain and cell loss, where the rate of proliferating mutant tumour cells exceeds the rate of those that die. Apoptosis, the most renowned form of programmed cell death, operates as a key physiological mechanism that limits cell population expansion, either to maintain tissue homeostasis or to remove potentially harmful cells, such as those that have sustained DNA damage. Paradoxically, high-grade cancers are generally associated with high constitutive levels of apoptosis. In cancer, cell-autonomous apoptosis constitutes a common tumour suppressor mechanism, a property which is exploited in cancer therapy. By contrast, limited apoptosis in the tumour-cell population also has the potential to promote cell survival and resistance to therapy by conditioning the tumour microenvironment (TME)-including phagocytes and viable tumour cells-and engendering pro-oncogenic effects. Notably, the constitutive apoptosis-mediated activation of cells of the innate immune system can help orchestrate a pro-oncogenic TME and may also effect evasion of cancer treatment. Here, we present an overview of the implications of cell death programmes in tumour biology, with particular focus on apoptosis as a process with "double-edged" consequences: on the one hand, being tumour suppressive through deletion of malignant or pre-malignant cells, while, on the other, being tumour progressive through stimulation of reparatory and regenerative responses in the TME.
Collapse
|
31
|
Abstract
The number of hair follicle stem cells decreases during aging and in hair-loss disorders, such as alopecia. In this issue of Cell Stem Cell, Xie et al. (2021) discover that the hair shaft serves as a physical niche component for the preservation of hair follicle stem cells.
Collapse
Affiliation(s)
- Taylor Hinnant
- Departments of Dermatology and Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Terry Lechler
- Departments of Dermatology and Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
32
|
Haddad A, Voth B, Brooks J, Swang M, Carryl H, Algarzae N, Taylor S, Parker C, Van Rompay KKA, De Paris K, Burke MW. Reduced neuronal population in the dorsolateral prefrontal cortex in infant macaques infected with simian immunodeficiency virus (SIV). J Neurovirol 2021; 27:923-935. [PMID: 34554407 PMCID: PMC8901521 DOI: 10.1007/s13365-021-01019-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/11/2021] [Accepted: 08/26/2021] [Indexed: 11/29/2022]
Abstract
Pediatric HIV infection remains a global health crisis with an estimated 150,000 new mother-to-child (MTCT) infections each year. Antiretroviral therapy (ART) has improved childhood survival, but only an estimated 53% of children worldwide have access to treatment. Adding to the health crisis is the neurological impact of HIV on the developing brain, in particular cognitive and executive function, which persists even when ART is available. Imaging studies suggest structural, connectivity, and functional alterations in perinatally HIV-infected youth. However, the paucity of histological data limits our ability to identify specific cortical regions that may underlie the clinical manifestations. Utilizing the pediatric simian immunodeficiency virus (SIV) infection model in infant macaques, we have previously shown that early-life SIV infection depletes the neuronal population in the hippocampus. Here, we expand on these previous studies to investigate the dorsolateral prefrontal cortex (dlPFC). A total of 11 ART-naïve infant rhesus macaques (Macaca mulatta) from previous studies were retrospectively analyzed. Infant macaques were either intravenously (IV) inoculated with highly virulent SIVmac251 at ~1 week of age and monitored for 6-10 weeks or orally challenged with SIVmac251 from week 9 of age onwards with a monitoring period of 10-23 weeks post-infection (19-34 weeks of age), and SIV-uninfected controls were euthanized at 16-17 weeks of age. Both SIV-infected groups show a significant loss of neurons along with evidence of ongoing neuronal death. Oral- and IV-infected animals showed a similar neuronal loss which was negatively correlated to chronic viremia levels as assessed by an area under the curve (AUC) analysis. The loss of dlPFC neurons may contribute to the rapid neurocognitive decline associated with pediatric HIV infection.
Collapse
Affiliation(s)
- Alexandra Haddad
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Brittany Voth
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Janiya Brooks
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Melanie Swang
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Heather Carryl
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Norah Algarzae
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
- King Saudi University, Riyadh, Riyadh, Kingdom of Saudi Arabia
| | - Shane Taylor
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Camryn Parker
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California Davis, Davis, CA, 95616, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Mark W Burke
- Department of Physiology and Biophysics, Howard University, Washington, DC, 20059, USA.
| |
Collapse
|
33
|
Karkali K, Martin-Blanco E. Dissection of the Regulatory Elements of the Complex Expression Pattern of Puckered, a Dual-Specificity JNK Phosphatase. Int J Mol Sci 2021; 22:ijms222212205. [PMID: 34830088 PMCID: PMC8623796 DOI: 10.3390/ijms222212205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/19/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
For developmental processes, we know most of the gene networks controlling specific cell responses. We still have to determine how these networks cooperate and how signals become integrated. The JNK pathway is one of the key elements modulating cellular responses during development. Yet, we still know little about how the core components of the pathway interact with additional regulators or how this network modulates cellular responses in the whole organism in homeostasis or during tissue morphogenesis. We have performed a promoter analysis, searching for potential regulatory sequences of puckered (puc) and identified different specific enhancers directing gene expression in different tissues and at different developmental times. Remarkably, some of these domains respond to the JNK activity, but not all. Altogether, these analyses show that puc expression regulation is very complex and that JNK activities participate in non-previously known processes during the development of Drosophila.
Collapse
|
34
|
Klemm J, Stinchfield MJ, Harris RE. Necrosis-induced apoptosis promotes regeneration in Drosophila wing imaginal discs. Genetics 2021; 219:6365941. [PMID: 34740246 PMCID: PMC8570793 DOI: 10.1093/genetics/iyab144] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/26/2021] [Indexed: 01/13/2023] Open
Abstract
Regeneration is a complex process that requires a coordinated genetic response to tissue loss. Signals from dying cells are crucial to this process and are best understood in the context of regeneration following programmed cell death, like apoptosis. Conversely, regeneration following unregulated forms of death, such as necrosis, have yet to be fully explored. Here, we have developed a method to investigate regeneration following necrosis using the Drosophila wing imaginal disc. We show that necrosis stimulates regeneration at an equivalent level to that of apoptosis-mediated cell death and activates a similar response at the wound edge involving localized JNK signaling. Unexpectedly, however, necrosis also results in significant apoptosis far from the site of ablation, which we have termed necrosis-induced apoptosis (NiA). This apoptosis occurs independent of changes at the wound edge and importantly does not rely on JNK signaling. Furthermore, we find that blocking NiA limits proliferation and subsequently inhibits regeneration, suggesting that tissues damaged by necrosis can activate programmed cell death at a distance from the injury to promote regeneration.
Collapse
Affiliation(s)
- Jacob Klemm
- School of Life Sciences, Arizona State University, Tempe, AZ 85728, USA
| | | | - Robin E Harris
- School of Life Sciences, Arizona State University, Tempe, AZ 85728, USA
| |
Collapse
|
35
|
Villars A, Levayer R. Collective effects in epithelial cell death and cell extrusion. Curr Opin Genet Dev 2021; 72:8-14. [PMID: 34626896 DOI: 10.1016/j.gde.2021.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/06/2021] [Accepted: 09/14/2021] [Indexed: 01/09/2023]
Abstract
Programmed cell death, notably apoptosis, is an essential guardian of tissue homeostasis and an active contributor of organ shaping. While the regulation of apoptosis has been mostly analysed in the framework of a cell autonomous process, recent works highlighted important collective effects which can tune cell elimination. This is particularly relevant for epithelial cell death, which requires fine coordination with the neighbours in order to maintain tissue sealing during cell expulsion. In this review, we will focus on the recent advances which outline the complex multicellular communications at play during epithelial cell death and cell extrusion. We will first focus on the new unanticipated functions of neighbouring cells during extrusion, discuss the contribution of distant neighbours, and finally highlight the complex feedbacks generated by cell elimination on neighbouring cell death.
Collapse
Affiliation(s)
- Alexis Villars
- Institut Pasteur, Université de Paris, CNRS UMR3738, Department of Developmental and Stem Cell Biology, F-75015 Paris, France; Sorbonne Université, Collège Doctoral, F75005 Paris, France
| | - Romain Levayer
- Institut Pasteur, Université de Paris, CNRS UMR3738, Department of Developmental and Stem Cell Biology, F-75015 Paris, France.
| |
Collapse
|
36
|
Xie Y, Chen D, Jiang K, Song L, Qian N, Du Y, Yang Y, Wang F, Chen T. Hair shaft miniaturization causes stem cell depletion through mechanosensory signals mediated by a Piezo1-calcium-TNF-α axis. Cell Stem Cell 2021; 29:70-85.e6. [PMID: 34624205 DOI: 10.1016/j.stem.2021.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 07/19/2021] [Accepted: 09/13/2021] [Indexed: 12/17/2022]
Abstract
In aging, androgenic alopecia, and genetic hypotrichosis disorders, hair shaft miniaturization is often associated with hair follicle stem cell (HFSC) loss. However, the mechanism causing this stem cell depletion in vivo remains elusive. Here we show that hair shaft loss or a reduction in diameter shrinks the physical niche size, which results in mechanical compression of HFSCs and their apoptotic loss. Mechanistically, cell compression activates the mechanosensitive channel Piezo1, which triggers calcium influx. This confers tumor necrosis factor alpha (TNF-α) sensitivity in a hair-cycle-dependent manner in otherwise resistant HFSCs and induces ectopic apoptosis. Persistent hair shaft miniaturization during aging and genetic hypotrichosis disorders causes long-term HFSC loss by inducing continuous ectopic apoptosis through Piezo1. Our results identify an unconventional role of the inert hair shaft structure as a functional niche component governing HFSC survival and reveal a mechanosensory axis that regulates physical-niche-atrophy-induced stem cell depletion in vivo.
Collapse
Affiliation(s)
- Yuhua Xie
- China Agricultural University, Beijing, China; National Institute of Biological Sciences, Beijing, China
| | - Daoming Chen
- National Institute of Biological Sciences, Beijing, China
| | - Kaiju Jiang
- National Institute of Biological Sciences, Beijing, China
| | - Lifang Song
- National Institute of Biological Sciences, Beijing, China
| | - Nannan Qian
- National Institute of Biological Sciences, Beijing, China
| | - Yingxue Du
- National Institute of Biological Sciences, Beijing, China
| | - Yong Yang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing, China
| | - Ting Chen
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
37
|
Soler Beatty J, Molnar C, Luque CM, de Celis JF, Martín-Bermudo MD. EGFRAP encodes a new negative regulator of the EGFR acting in both normal and oncogenic EGFR/Ras-driven tissue morphogenesis. PLoS Genet 2021; 17:e1009738. [PMID: 34411095 PMCID: PMC8407591 DOI: 10.1371/journal.pgen.1009738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 08/31/2021] [Accepted: 07/23/2021] [Indexed: 12/27/2022] Open
Abstract
Activation of Ras signaling occurs in ~30% of human cancers. However, activated Ras alone is insufficient to produce malignancy. Thus, it is imperative to identify those genes cooperating with activated Ras in driving tumoral growth. In this work, we have identified a novel EGFR inhibitor, which we have named EGFRAP, for EGFR adaptor protein. Elimination of EGFRAP potentiates activated Ras-induced overgrowth in the Drosophila wing imaginal disc. We show that EGFRAP interacts physically with the phosphorylated form of EGFR via its SH2 domain. EGFRAP is expressed at high levels in regions of maximal EGFR/Ras pathway activity, such as at the presumptive wing margin. In addition, EGFRAP expression is up-regulated in conditions of oncogenic EGFR/Ras activation. Normal and oncogenic EGFR/Ras-mediated upregulation of EGRAP levels depend on the Notch pathway. We also find that elimination of EGFRAP does not affect overall organogenesis or viability. However, simultaneous downregulation of EGFRAP and its ortholog PVRAP results in defects associated with increased EGFR function. Based on these results, we propose that EGFRAP is a new negative regulator of the EGFR/Ras pathway, which, while being required redundantly for normal morphogenesis, behaves as an important modulator of EGFR/Ras-driven tissue hyperplasia. We suggest that the ability of EGFRAP to functionally inhibit the EGFR pathway in oncogenic cells results from the activation of a feedback loop leading to increase EGFRAP expression. This could act as a surveillance mechanism to prevent excessive EGFR activity and uncontrolled cell growth. Activation of Ras signalling occurs in ~30% of human cancers. However, activated Ras alone is insufficient to produce malignancy. Thus, the discovery of genes cooperating with Ras in cancer is imperative to understand tumoral growth driven by Ras activating mutations. A key output of over-activated EGFR/Ras signalling is the induction of a complex and dynamic set of transcriptional networks leading to changes in gene expression. As a result of these changes, the normal function of some genes can become adjusted in a tumorigenic context. In this work, using the Drosophila wing imaginal disc as model system, we have identified a new EGFR inhibitor, EGFRAP, which function is redundant for proper morphogenesis, yet becomes an important limiter of the overgrowth driven by oncogenic EGFR/Ras activity. We show that the specificity of EGFRAP in cells with high levels of EGFR activity arises from activation of a negative feedback loop resulting in increased EGFRAP levels. This could act to prevent excessive EGFR activity and uncontrolled cell growth. We believe the identification of other factors behaving like EGFRAP, will help in our fight against cancer, as it might lead to the identification of new therapeutic drugs affecting cancer but not normal cells, a top priority in cancer research.
Collapse
Affiliation(s)
- Jennifer Soler Beatty
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC/JA, Sevilla, Spain
| | - Cristina Molnar
- Centro de Biología Molecular Severo Ochoa (UAM/CSIC), Univ. Autónoma de Madrid, Madrid, Spain
| | - Carlos M. Luque
- Centro de Biología Molecular Severo Ochoa (UAM/CSIC), Univ. Autónoma de Madrid, Madrid, Spain
| | - Jose F. de Celis
- Centro de Biología Molecular Severo Ochoa (UAM/CSIC), Univ. Autónoma de Madrid, Madrid, Spain
| | - María D. Martín-Bermudo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC/JA, Sevilla, Spain
- * E-mail:
| |
Collapse
|
38
|
Fu Y, Sui B, Xiang L, Yan X, Wu D, Shi S, Hu X. Emerging understanding of apoptosis in mediating mesenchymal stem cell therapy. Cell Death Dis 2021; 12:596. [PMID: 34108448 PMCID: PMC8190192 DOI: 10.1038/s41419-021-03883-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cell transplantation (MSCT) has been recognized as a potent and promising approach to achieve immunomodulation and tissue regeneration, but the mechanisms of how MSCs exert therapeutic effects remain to be elucidated. Increasing evidence suggests that transplanted MSCs only briefly remain viable in recipients, after which they undergo apoptosis in the host circulation or in engrafted tissues. Intriguingly, apoptosis of infused MSCs has been revealed to be indispensable for their therapeutic efficacy, while recipient cells can also develop apoptosis as a beneficial response in restoring systemic and local tissue homeostasis. It is notable that apoptotic cells produce apoptotic extracellular vesicles (apoEVs), traditionally known as apoptotic bodies (apoBDs), which possess characterized miRnomes and proteomes that contribute to their specialized function and to intercellular communication. Importantly, it has been demonstrated that the impact of apoEVs is long-lasting in health and disease contexts, and they critically mediate the efficacy of MSCT. In this review, we summarize the emerging understanding of apoptosis in mediating MSCT, highlighting the potential of apoEVs as cell-free therapeutics.
Collapse
Affiliation(s)
- Yu Fu
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, 350117, China.,South China Center of Craniofacial Stem Cell Research, Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Bingdong Sui
- South China Center of Craniofacial Stem Cell Research, Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China.,Research and Development Center for Tissue Engineering, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Lei Xiang
- South China Center of Craniofacial Stem Cell Research, Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Xutong Yan
- South China Center of Craniofacial Stem Cell Research, Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Di Wu
- South China Center of Craniofacial Stem Cell Research, Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China
| | - Songtao Shi
- South China Center of Craniofacial Stem Cell Research, Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, China.
| | - Xuefeng Hu
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, 350117, China.
| |
Collapse
|
39
|
Gagliardi PA, Dobrzyński M, Jacques MA, Dessauges C, Ender P, Blum Y, Hughes RM, Cohen AR, Pertz O. Collective ERK/Akt activity waves orchestrate epithelial homeostasis by driving apoptosis-induced survival. Dev Cell 2021; 56:1712-1726.e6. [PMID: 34081908 DOI: 10.1016/j.devcel.2021.05.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 02/16/2021] [Accepted: 05/09/2021] [Indexed: 12/20/2022]
Abstract
Cell death events continuously challenge epithelial barrier function yet are crucial to eliminate old or critically damaged cells. How such apoptotic events are spatio-temporally organized to maintain epithelial homeostasis remains unclear. We observe waves of extracellular-signal-regulated kinase (ERK) and AKT serine/threonine kinase (Akt) activity pulses that originate from apoptotic cells and propagate radially to healthy surrounding cells. This requires epidermal growth factor receptor (EGFR) and matrix metalloproteinase (MMP) signaling. At the single-cell level, ERK/Akt waves act as spatial survival signals that locally protect cells in the vicinity of the epithelial injury from apoptosis for a period of 3-4 h. At the cell population level, ERK/Akt waves maintain epithelial homeostasis (EH) in response to mild or intense environmental insults. Disruption of this spatial signaling system results in the inability of a model epithelial tissue to ensure barrier function in response to environmental insults.
Collapse
Affiliation(s)
| | - Maciej Dobrzyński
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Marc-Antoine Jacques
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Coralie Dessauges
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Pascal Ender
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Yannick Blum
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Robert M Hughes
- Department of Chemistry, East Carolina University, 300 Science and Technology Building, Greenville, NC 27858-4353, USA
| | - Andrew R Cohen
- Department of Electrical and Computer Engineering, Drexel University, 3120-40 Market Street, Suite 313, Philadelphia, PA 19104, USA
| | - Olivier Pertz
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland.
| |
Collapse
|
40
|
Wang FH, Zhang L, Gong G, Yan XC, Zhang LT, Zhang FT, Liu HF, Lv Q, Wang ZY, Wang RJ, Zhang YJ, Wang ZX, Liu ZH, He LB, Su R, Zhao YH, Li JQ. Genome-wide association study of fleece traits in Inner Mongolia Cashmere goats. Anim Genet 2021; 52:375-379. [PMID: 33778967 PMCID: PMC8251931 DOI: 10.1111/age.13053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2021] [Indexed: 12/14/2022]
Abstract
Inner Mongolia Cashmere goat is a well-known local cashmere goat breed in China. It is famous for excellent fleece quality and a significant advantage in cashmere yield compared to other cashmere goat breeds. In this study, a genome-wide association study was used to investigate fiber length, fiber diameter, and cashmere yield of 192 Inner Mongolia Cashmere goats using the Illumina GoatSNP52K Beadchip panel. We discovered that four single nucleotide polymorphisms (SNPs) reached genome-wide significance levels. These SNPs were located in some genes, e.g. FGF12, SEMA3D, EVPL, and SOX5, possibly related to fleece traits in Inner Mongolia Cashmere goat. Gene ontology enrichment analysis revealed that these genes were enriched in several biological pathways that were involved in hair follicle development in cashmere goats. In summary, the identified significant SNPs and genes provide useful information to explore genetic mechanisms underlying the variation in fleece traits and genomic selection of Chinese cashmere goat.
Collapse
Affiliation(s)
- F. H. Wang
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - L. Zhang
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - G. Gong
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - X. C. Yan
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - L. T. Zhang
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - F. T. Zhang
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - H. F. Liu
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - Q. Lv
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - Z. Y. Wang
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - R. J. Wang
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - Y. J. Zhang
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - Z. X. Wang
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
- Key Laboratory of Animal Genetics, Breeding and ReproductionHohhotInner Mongolia Autonomous Region010018China
| | - Z. H. Liu
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
- Key Laboratory of Mutton Sheep Genetics and BreedingMinistry of Agriculture and Rural AffairsHohhot010018China
| | - L. B. He
- Key Laboratory of Mutton Sheep Genetics and BreedingMinistry of Agriculture and Rural AffairsHohhot010018China
| | - R. Su
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - Y. H. Zhao
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| | - J. Q. Li
- College of Animal ScienceInner Mongolia Agricultural UniversityHohhotInner Mongolia010018China
| |
Collapse
|
41
|
Herrera SC, Bach EA. The Emerging Roles of JNK Signaling in Drosophila Stem Cell Homeostasis. Int J Mol Sci 2021; 22:ijms22115519. [PMID: 34073743 PMCID: PMC8197226 DOI: 10.3390/ijms22115519] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
The Jun N-terminal kinase (JNK) pathway is an evolutionary conserved kinase cascade best known for its roles during stress-induced apoptosis and tumor progression. Recent findings, however, have identified new roles for this pleiotropic pathway in stem cells during regenerative responses and in cellular plasticity. Here, we provide an overview of recent findings about the new roles of JNK signaling in stem cell biology using two well-established Drosophila models: the testis and the intestine. We highlight the pathway’s roles in processes such as proliferation, death, self-renewal and reprogramming, and discuss the known parallels between flies and mammals.
Collapse
Affiliation(s)
- Salvador C. Herrera
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Carretera de Utrera km 1, 41018 Sevilla, Spain
- Correspondence: (S.C.H.); (E.A.B.)
| | - Erika A. Bach
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Correspondence: (S.C.H.); (E.A.B.)
| |
Collapse
|
42
|
Abstract
Drosophila melanogaster remains a foremost genetic model to study basic cell biological processes in the context of multi-cellular development. In such context, the behavior of one cell can influence another. Non-autonomous signaling among cells occurs throughout metazoan development and disease, and is too vast to be covered by a single review. I will focus here on non-autonomous signaling events that occur in response to cell death in the larval epithelia and affect the life-death decision of surviving cells. I will summarize the use of Drosophila to study cell death-induced proliferation, apoptosis-induced apoptosis, and apoptosis-induced survival signaling. Key insights from Drosophila will be discussed in the context of analogous processes in mammalian development and cancer biology.
Collapse
Affiliation(s)
- Tin Tin Su
- Department of Molecular, Cellular and Developmental Biology, 347 UCB, University of Colorado, Boulder, CO 80309-0347, USA
| |
Collapse
|
43
|
Wang X, Liang H, Xu W, Ma X. Wallenda-Nmo Axis Regulates Growth via Hippo Signaling. Front Cell Dev Biol 2021; 9:658288. [PMID: 33937258 PMCID: PMC8085559 DOI: 10.3389/fcell.2021.658288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/04/2021] [Indexed: 12/26/2022] Open
Abstract
Both Hippo signaling pathways and cell polarity regulation are critical for cell proliferation and the maintenance of tissue homeostasis, despite the well-established connections between cell polarity disruption and Hippo inactivation, the molecular mechanism by which aberrant cell polarity induces Hippo-mediated overgrowth remains underexplored. Here we use Drosophila wing discs as a model and identify the Wnd-Nmo axis as an important molecular link that bridges loss-of-cell polarity-triggered Hippo inactivation and overgrowth. We show that Wallenda (Wnd), a MAPKKK (mitogen-activated protein kinase kinase kinase) family member, is a novel regulator of Hippo pathways in Drosophila and that overexpression of Wnd promotes growth via Nemo (Nmo)- mediated Hippo pathway inactivation. We further demonstrate that both Wnd and Nmo are required for loss-of-cell polarity-induced overgrowth and Hippo inactivation. In summary, our findings provide a novel insight on how cell polarity loss contributes to overgrowth and uncover the Wnd-Nmo axis as an essential additional branch that regulates Hippo pathways in Drosophila.
Collapse
Affiliation(s)
- Xianping Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.,Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Hui Liang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.,Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Wenyan Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.,Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China.,Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xianjue Ma
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.,Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| |
Collapse
|
44
|
Guntur AR, Venkatanarayan A, Gangula S, Lundell MJ. Zfh-2 facilitates Notch-induced apoptosis in the CNS and appendages of Drosophila melanogaster. Dev Biol 2021; 475:65-79. [PMID: 33705738 DOI: 10.1016/j.ydbio.2021.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 12/26/2022]
Abstract
Apoptosis is a fundamental remodeling process for most tissues during development. In this manuscript we examine a pro-apoptotic function for the Drosophila DNA binding protein Zfh-2 during development of the central nervous system (CNS) and appendages. In the CNS we find that a loss-of-function zfh-2 allele gives an overall reduction of apoptotic cells in the CNS, and an altered pattern of expression for the axonal markers 22C10 and FasII. This same loss-of-function zfh-2 allele causes specific cells in the NB7-3 lineage of the CNS that would normally undergo apoptosis to be inappropriately maintained, whereas a gain-of-function zfh-2 allele has the opposite effect, resulting in a loss of normal NB 7-3 progeny. We also demonstrate that Zfh-2 and Hunchback reciprocally repress each other's gene expression which limits apoptosis to later born progeny of the NB7-3 lineage. Apoptosis is also required for proper segmentation of the fly appendages. We find that Zfh-2 co-localizes with apoptotic cells in the folds of the imaginal discs and presumptive cuticular joints. A reduction of Zfh-2 levels with RNAi inhibits expression of the pro-apoptotic gene reaper, and produces abnormal joints in the leg, antenna and haltere. Apoptosis has previously been shown to be activated by Notch signaling in both the NB7-3 CNS lineage and the appendage joints. Our results indicate that Zfh-2 facilitates Notch-induced apoptosis in these structures.
Collapse
Affiliation(s)
- Ananya R Guntur
- Department of Biology, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | | | - Sindhura Gangula
- Department of Biology, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Martha J Lundell
- Department of Biology, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| |
Collapse
|
45
|
Hao X, Li W. Chloroquine diphosphate suppresses liver cancer via inducing apoptosis in Wistar rats using interventional therapy. Oncol Lett 2021; 21:233. [PMID: 33613722 DOI: 10.3892/ol.2021.12494] [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: 07/20/2020] [Accepted: 12/08/2020] [Indexed: 11/05/2022] Open
Abstract
Liver cancer ranks as the second leading cause of cancer-associated mortality worldwide. To date, neither current ablation therapy nor chemotherapy are considered ideal in improving the outcome of liver cancer. Therefore, more effective therapies for treating this devastating disease are urgently required. Interventional therapy has been used for numerous years in the treatment of different types of cancer, and is characterized by the direct delivery of anticancer drugs into the tumor. It has been reported that antimalarial chloroquine diphosphate (CQ) exerts effective anticancer activity against several types of cancer. However, its effect on liver cancer remains unclear. Therefore, in the present study, 2D monolayer cell culture and 3D spheroid in vitro models, and a rat model, were utilized to investigate the effect of CQ on liver cancer. CQ demonstrated an effective anticancer effect on HepG2 cells and 3D liver spheroids. Furthermore, the drug significantly inhibited cell growth and viability in the 2D and 3D in vitro models. The CQ-based intervention treatment effectively attenuated tumor size and weight, increased food intake and consumption of drinking water, and improved body weight and survival rate of rats in the in vivo model. In addition, treatment with CQ potently increased the expression levels of the apoptosis-related genes. Taken together, the findings of the present study may provide a novel insight into the development of safe and effective treatments for liver cancer.
Collapse
Affiliation(s)
- Xiaoguang Hao
- Department of Radiology, The Fourth Hospital of Hebei Medicine University, Shijiazhuang, Hebei 050000, P.R. China
| | - Weijing Li
- Department of Anesthesiology, The Fourth Hospital of Hebei Medicine University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
46
|
Koren E, Fuchs Y. Modes of Regulated Cell Death in Cancer. Cancer Discov 2021; 11:245-265. [PMID: 33462123 DOI: 10.1158/2159-8290.cd-20-0789] [Citation(s) in RCA: 199] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/15/2020] [Accepted: 10/29/2020] [Indexed: 11/16/2022]
Abstract
Cell suicide pathways, termed regulated cell death (RCD), play a critical role in organismal development, homeostasis, and pathogenesis. Here, we provide an overview of key RCD modalities, namely apoptosis, entosis, necroptosis, pyroptosis, and ferroptosis. We explore how various RCD modules serve as a defense mechanism against the emergence of cancer as well as the manner in which they can be exploited to drive oncogenesis. Furthermore, we outline current therapeutic agents that activate RCD and consider novel RCD-based strategies for tumor elimination. SIGNIFICANCE: A variety of antitumor therapeutics eliminate cancer cells by harnessing the devastating potential of cellular suicide pathways, emphasizing the critical importance of RCD in battling cancer. This review supplies a mechanistic perspective of distinct RCD modalities and explores the important role they play in tumorigenesis. We discuss how RCD modules serve as a double-edged sword as well as novel approaches aimed at selectively manipulating RCD for tumor eradication.
Collapse
Affiliation(s)
- Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel. Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel. Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
47
|
Clustered rapid induction of apoptosis limits ZIKV and DENV-2 proliferation in the midguts of Aedes aegypti. Commun Biol 2021; 4:69. [PMID: 33452408 PMCID: PMC7810730 DOI: 10.1038/s42003-020-01614-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 12/15/2020] [Indexed: 11/29/2022] Open
Abstract
Inter-host transmission of pathogenic arboviruses such as dengue virus (DENV) and Zika virus (ZIKV) requires systemic infection of the mosquito vector. Successful systemic infection requires initial viral entry and proliferation in the midgut cells of the mosquito followed by dissemination to secondary tissues and eventual entry into salivary glands1. Lack of arbovirus proliferation in midgut cells has been observed in several Aedes aegypti strains2, but the midgut antiviral responses underlying this phenomenon are not yet fully understood. We report here that there is a rapid induction of apoptosis (RIA) in the Aedes aegypti midgut epithelium within 2 hours of infection with DENV-2 or ZIKV in both in vivo blood-feeding and ex vivo midgut infection models. Inhibition of RIA led to increased virus proliferation in the midgut, implicating RIA as an innate immune mechanism mediating midgut infection in this mosquito vector. Ayers et al. report rapid induction of apoptosis in the midgut of Aedes aegypti mosquitoes within 2 hours of infection by dengue and Zika viruses, and find that inhibiting apoptosis led to increased virus proliferation in the midgut. These results suggest rapid induction of apoptosis as an innate immune mechanism mediating midgut infection in this mosquito vector.
Collapse
|
48
|
Kumar R, Saneja A, Panda AK. An Annexin V-FITC-Propidium Iodide-Based Method for Detecting Apoptosis in a Non-Small Cell Lung Cancer Cell Line. Methods Mol Biol 2021; 2279:213-223. [PMID: 33683697 DOI: 10.1007/978-1-0716-1278-1_17] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Annexin V and propidium iodide staining is widely used for determining the cellular death through apoptosis. In the presence of Ca2+ ions, annexin V has a strong binding affinity for phosphatidylserine, a membrane phospholipid that during apoptosis is translocated from the inner side of the cell membrane to its outer side. On the other hand, propidium iodide has ability for DNA binding and it can only enter into necrotic or late apoptotic cells. This chapter describes a commonly used method for detection of apoptosis in a non-small cell lung cancer cell line using annexin V and propidium iodide dye. We describe the detection of different stages of apoptosis in the A549 lung cancer cell line treated with dihydroartemisinin (DHA). This apoptosis detection method can be used to determine the efficacy of different kinds of drugs on cultured cancer cell lines.
Collapse
Affiliation(s)
- Robin Kumar
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Ankit Saneja
- Product Development Cell, National Institute of Immunology, New Delhi, India.
| | - Amulya K Panda
- Product Development Cell, National Institute of Immunology, New Delhi, India.
| |
Collapse
|
49
|
Sharma V, Mutsuddi M, Mukherjee A. Deltex cooperates with TRAF6 to promote apoptosis and cell migration through Eiger-independent JNK activation in Drosophila. Cell Biol Int 2020; 45:686-700. [PMID: 33300258 DOI: 10.1002/cbin.11521] [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: 07/24/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/20/2022]
Abstract
JNK signaling is a highly conserved signaling pathway that regulates a broad spectrum of cellular processes including cell proliferation, migration, and apoptosis. In Drosophila, JNK signaling is activated by binding of the tumor necrosis factor (TNF) Eiger to its receptor Wengen, and a conserved signaling cascade operates that culminates into activation of dual phosphatase Puckered thereby triggering apoptosis. The tumor necrosis factor receptor (TNFR) associated factor 6 (TRAF6) is an adaptor protein, which transduces the signal from TNFRs and Toll-like receptor/interleukin-1 receptor superfamily to induce a wide spectrum of cellular responses. TRAF6 also acts as the adaptor protein that mediates Eiger/JNK signaling in Drosophila. In a genetic interaction study, deltex (Dx) was identified as a novel interactor of TRAF6. Dx is well known to regulate Notch signaling in a context-dependent manner. Our data suggest that combinatorial action of Dx and TRAF6 enhances the Dx-induced wing nicking phenotype by inducing caspase-mediated cell death. Co-expression of Dx and TRAF6 also results in enhanced invasive behavior and perturbs the normal morphology of cells. The cooperative action of Dx and TRAF6 is attributed to JNK activation, which also leads to ectopic wingless (Wg) and decapentaplegic (Dpp) expression. Our results also reveal that the endocytic pathway component Rab7 may play a pivotal role in the regulation of Dx-TRAF6-mediated activation of JNK signaling. Here, we present the fact that Dx and TRAF6 together activate JNK signaling in an Eiger-independent mechanism.
Collapse
Affiliation(s)
- Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
50
|
Domesticated and optimized mitochondria: Mitochondrial modifications based on energetic status and cellular stress. Life Sci 2020; 265:118766. [PMID: 33245965 DOI: 10.1016/j.lfs.2020.118766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/28/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria are the main source of energy and play an important role in coupling intracellular and intercellular metabolic cooperation. Cellular stress and energetic status can affect various mitochondrial behaviors, including mitochondrial biogenesis, mitophagy, assembly of respiratory chain supercomplexes and mitochondrial distribution. These modifications usually result in adaptive adjustment of mitochondrial output and resistance to cellular stress. However, when the pro-death signals triggered by excessive damage converge to mitochondria, mitochondrial reserve and functional status can profoundly determine the direction of cell death, and even affect the survival and death of surrounding or distant tissues. In this review, we discuss multiple mitochondrial modifications in eukaryotes based on metabolic status and cellular stress, and review the emerging knowledge about the effects of mitochondrial dysfunction on the fate of cells and surrounding tissues.
Collapse
|