1
|
Fischer Z, Nauman C, Shayestehpour S, Pence L, Bouyain S, Yao X, Dobens LL. The Drosophila pseudokinase Tribbles translocates to the fat body membrane in response to fasting to modulate insulin sensitivity. Development 2025; 152:dev204493. [PMID: 40292740 PMCID: PMC12070071 DOI: 10.1242/dev.204493] [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: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 04/30/2025]
Abstract
The Drosophila pseudokinase Tribbles (Trbl) shares conserved functions with human TRIB3 to bind and inhibit Akt phosphorylation-activation by the Insulin Receptor (InR) to reduce insulin responses; consistent with this, increased levels of human TRIB3 are linked to type 2 diabetes. Here, we show that in fat body cells of well-fed Drosophila larvae, Trbl expression is low and predominantly in the nucleus while fasting or genetic reduction of insulin signaling resulted in increased Trbl expression and Trbl protein translocation to the plasma membrane. An E/G mutation in the Trbl pseudokinase kinase activation loop dominantly interfered with Trbl function leading to increased Akt activity, increased stability of Trbl substrates, including Trbl itself, and aberrant redistribution of Trbl multimers to the membrane. Several strategies designed to increase Akt activity were sufficient to translocate Trbl to the membrane, consistent with the notion that subcellular trafficking of Trbl to the fat body cell membrane acts a rheostat to reduce the strength of Akt-mediated insulin responses, counter to the InR, which has been shown to redistribute away from the membrane to modulate insulin signaling.
Collapse
Affiliation(s)
- Zachary Fischer
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Christopher Nauman
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Shima Shayestehpour
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Laramie Pence
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Samuel Bouyain
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Xiaolan Yao
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Leonard L. Dobens
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| |
Collapse
|
2
|
Cruz J, Sun WY, Verbeke A, Hariharan IK. Single-cell transcriptomics of X-ray irradiated Drosophila wing discs reveals heterogeneity related to cell-cycle status and cell location. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.10.627868. [PMID: 39990483 PMCID: PMC11844406 DOI: 10.1101/2024.12.10.627868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Even seemingly homogeneous populations of cells can express phenotypic diversity in response to environmental changes. Thus, X-ray irradiation of tissues composed of diverse cell types can have complex outcomes. We have used single-cell RNA-sequencing to study the effects of X-ray radiation on the Drosophila wing imaginal disc, a relatively simple tissue composed mostly of epithelial cells. Transcriptomic clustering of cells collected from the wing disc generates clusters that are mainly grouped based on proximodistal cell location. To quantify heterogeneity of gene expression among clusters, we adapted a metric used to study market concentration, the Herfindahl-Hirschman Index. Genes involved in DNA damage repair, defense against reactive oxygen species, cell cycle progression, and apoptosis are expressed relatively uniformly. In contrast, genes encoding a subset of ligands, notably cytokines that activate the JAK/STAT pathway, some transcription factors including Ets21C, previously implicated in regeneration, and several signaling proteins are expressed more regionally. Though the radiation-responsive transcription factor p53 is expressed relatively uniformly in the wing disc, several regionally-induced genes still require p53 function, indicating that regional and radiation-induced factors combine to regulate their expression. We also examined heterogeneity within regions using a clustering approach based on cell cycle gene expression. A subpopulation of cells, characterized by high levels of tribbles expression, is amplified in irradiated discs. Remarkably, this subpopulation accounts for a considerable fraction of radiation-induced gene expression, indicating that cellular responses are non-uniform even within regions. Thus, both inter-regional and intra-regional heterogeneity are important features of tissue responses to X-ray radiation.
Collapse
Affiliation(s)
- Joyner Cruz
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - Willam Y. Sun
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - Alexandra Verbeke
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - Iswar K. Hariharan
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| |
Collapse
|
3
|
Sabbagh S, Zhang H, Harris TJC. Drosophila anterior midgut internalization via collective epithelial-mesenchymal transition at the embryo surface and enclosure by surrounding tissues. Dev Biol 2025; 517:191-202. [PMID: 39393484 DOI: 10.1016/j.ydbio.2024.10.002] [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: 08/13/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Internal organ development requires cell internalization, which can occur individually or collectively. The best characterized mode of collective internalization is epithelial invagination. Alternate modes involving collective mesenchymal behaviours at the embryo surface have been documented, but their prevalence is unclear. The Drosophila embryo has been a major model for the study of epithelial invaginations. However, internalization of the Drosophila anterior midgut primordium is incompletely understood. Here, we report that an epithelial-mesenchymal transition (EMT) occurs across the internalizing primordium when it is still at the embryo surface. At the earliest internalization stage, the primordium displays less junctional DE-cadherin than surrounding tissues but still exhibits coordinated epithelial structure as it invaginates with the ventral furrow. This initial invagination is transient, and its loss correlates with the activation of an associated mitotic domain. Activation of a subsequent mitotic domain across the broader primordium results in cell divisions with mixed orientations that deposit some cells within the embryo. However, cell division is non-essential for primordium internalization. Post-mitotically, the surface primordium displays hallmarks of EMT: loss of adherens junctions, loss of epithelial cell polarity, and gain of cell protrusions. Primordium cells extend over each other as they internalize asynchronously as individuals or small groups, and the primordium becomes enclosed by the reorganizations of surrounding epithelial tissues. We propose that collective EMT at the embryo surface promotes anterior midgut internalization through both inwardly-directed divisions and movements of its cells, and that the latter process is facilitated by surrounding tissue remodeling.
Collapse
Affiliation(s)
- Sandra Sabbagh
- Department of Cell & Systems Biology, University of Toronto, Canada
| | - Hui Zhang
- Department of Cell & Systems Biology, University of Toronto, Canada
| | - Tony J C Harris
- Department of Cell & Systems Biology, University of Toronto, Canada.
| |
Collapse
|
4
|
Yeo H, Lim JH, Eom J, Kim M, Kwon H, Kang SW, Song Y. Diet-induced obesity and aging-induced upregulation of Trib3 interfere with energy homeostasis by downregulating the thermogenic capacity of BAT. Exp Mol Med 2024; 56:2690-2702. [PMID: 39623091 DOI: 10.1038/s12276-024-01361-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/30/2024] [Accepted: 09/24/2024] [Indexed: 12/28/2024] Open
Abstract
Characterized by UCP1 expression and abundant mitochondria, brown adipose tissue (BAT) plays a crucial role in energy balance by converting chemical energy into heat through the cost of ATP production. In this study, it was demonstrated that Trib3 is a critical determinant of BAT-mediated energy expenditure and whole-body energy homeostasis. Under 60% high-fat diet conditions, Trib3 expression in BAT was elevated. Mice deficient in Trib3 are resistant to diet-induced obesity and exhibit improved glucose homeostasis due to enhanced BAT activity. Furthermore, brown adipocyte progenitor cells (APCs) lacking Trib3 exhibited increased proliferation and promoted brown adipocyte differentiation and mitochondrial biogenesis, contributing to the increase in the maximal thermogenic capacity of BAT in Trib3-deficient mice. Mechanistically, it was discovered that Trib3 expression is upregulated by free fatty acids at the transcriptional level and synergistically upregulated by DAG-PKC at the posttranslational level. This occurs through the modulation of COP1-mediated Trib3 protein turnover. Interestingly, the level of Trib3 expression in BAT increased with age. Trib3 knockout mice were protected from aging-related weight gain and impaired glucose homeostasis. These results suggest that Trib3 acts as an obesity- and aging-associated factor that negatively regulates BAT activity and that the loss of Trib3 may provide a beneficial approach to prevent obesity and aging-associated metabolic syndrome by increasing the thermogenic capacity of BAT.
Collapse
Affiliation(s)
- Hyejin Yeo
- Department of Brain Science, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-Hye Lim
- Department of Brain Science, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji Eom
- Department of Brain Science, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - MinJeong Kim
- Department of Brain Science, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyeji Kwon
- Department of Brain Science, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Wook Kang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Youngsup Song
- Department of Brain Science, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
5
|
Hammond FR, Lewis A, Pollara G, Tomlinson GS, Noursadeghi M, Kiss-Toth E, Elks PM. Tribbles1 is host protective during in vivo mycobacterial infection. eLife 2024; 13:e95980. [PMID: 38896446 PMCID: PMC11186633 DOI: 10.7554/elife.95980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Tuberculosis is a major global health problem and is one of the top 10 causes of death worldwide. There is a pressing need for new treatments that circumvent emerging antibiotic resistance. Mycobacterium tuberculosis parasitises macrophages, reprogramming them to establish a niche in which to proliferate, therefore macrophage manipulation is a potential host-directed therapy if druggable molecular targets could be identified. The pseudokinase Tribbles1 (Trib1) regulates multiple innate immune processes and inflammatory profiles making it a potential drug target in infections. Trib1 controls macrophage function, cytokine production, and macrophage polarisation. Despite wide-ranging effects on leukocyte biology, data exploring the roles of Tribbles in infection in vivo are limited. Here, we identify that human Tribbles1 is expressed in monocytes and is upregulated at the transcript level after stimulation with mycobacterial antigen. To investigate the mechanistic roles of Tribbles in the host response to mycobacteria in vivo, we used a zebrafish Mycobacterium marinum (Mm) infection tuberculosis model. Zebrafish Tribbles family members were characterised and shown to have substantial mRNA and protein sequence homology to their human orthologues. trib1 overexpression was host-protective against Mm infection, reducing burden by approximately 50%. Conversely, trib1 knockdown/knockout exhibited increased infection. Mechanistically, trib1 overexpression significantly increased the levels of proinflammatory factors il-1β and nitric oxide. The host-protective effect of trib1 was found to be dependent on the E3 ubiquitin kinase Cop1. These findings highlight the importance of Trib1 and Cop1 as immune regulators during infection in vivo and suggest that enhancing macrophage TRIB1 levels may provide a tractable therapeutic intervention to improve bacterial infection outcomes in tuberculosis.
Collapse
Affiliation(s)
- Ffion R Hammond
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Amy Lewis
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Gabriele Pollara
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Gillian S Tomlinson
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Mahdad Noursadeghi
- Division of Infection & Immunity, University College LondonLondonUnited Kingdom
| | - Endre Kiss-Toth
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| | - Philip M Elks
- The Bateson Centre, School of Medicine and Population Health, Faculty of Health, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
6
|
Singh K, Showalter CA, Manring HR, Haque SJ, Chakravarti A. "Oh, Dear We Are in Tribble": An Overview of the Oncogenic Functions of Tribbles 1. Cancers (Basel) 2024; 16:1889. [PMID: 38791967 PMCID: PMC11120034 DOI: 10.3390/cancers16101889] [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: 04/18/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Pseudokinases are catalytically inactive proteins in the human genome that lack the ability to transfer phosphate from ATP to their substrates. The Tribbles family of pseudokinases contains three members: Tribbles 1, 2, and 3. Tribbles 1 has recently gained importance because of its involvement in various diseases, including cancer. It acts as a scaffolding protein that brings about the degradation of its substrate proteins, such as C/EBPα/β, MLXIPL, and RAR/RXRα, among others, via the ubiquitin proteasome system. It also serves as an adapter protein, which sequesters different protein molecules and activates their downstream signaling, leading to processes, such as cell survival, cell proliferation, and lipid metabolism. It has been implicated in cancers such as AML, prostate cancer, breast cancer, CRC, HCC, and glioma, where it activates oncogenic signaling pathways such as PI3K-AKT and MAPK and inhibits the anti-tumor function of p53. TRIB1 also causes treatment resistance in cancers such as NSCLC, breast cancer, glioma, and promyelocytic leukemia. All these effects make TRIB1 a potential drug target. However, the lack of a catalytic domain renders TRIB1 "undruggable", but knowledge about its structure, conformational changes during substrate binding, and substrate binding sites provides an opportunity to design small-molecule inhibitors against specific TRIB1 interactions.
Collapse
Affiliation(s)
| | | | | | | | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
7
|
Zhang SX, Wang JJ, Starr CR, Lee EJ, Park KS, Zhylkibayev A, Medina A, Lin JH, Gorbatyuk M. The endoplasmic reticulum: Homeostasis and crosstalk in retinal health and disease. Prog Retin Eye Res 2024; 98:101231. [PMID: 38092262 PMCID: PMC11056313 DOI: 10.1016/j.preteyeres.2023.101231] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The endoplasmic reticulum (ER) is the largest intracellular organelle carrying out a broad range of important cellular functions including protein biosynthesis, folding, and trafficking, lipid and sterol biosynthesis, carbohydrate metabolism, and calcium storage and gated release. In addition, the ER makes close contact with multiple intracellular organelles such as mitochondria and the plasma membrane to actively regulate the biogenesis, remodeling, and function of these organelles. Therefore, maintaining a homeostatic and functional ER is critical for the survival and function of cells. This vital process is implemented through well-orchestrated signaling pathways of the unfolded protein response (UPR). The UPR is activated when misfolded or unfolded proteins accumulate in the ER, a condition known as ER stress, and functions to restore ER homeostasis thus promoting cell survival. However, prolonged activation or dysregulation of the UPR can lead to cell death and other detrimental events such as inflammation and oxidative stress; these processes are implicated in the pathogenesis of many human diseases including retinal disorders. In this review manuscript, we discuss the unique features of the ER and ER stress signaling in the retina and retinal neurons and describe recent advances in the research to uncover the role of ER stress signaling in neurodegenerative retinal diseases including age-related macular degeneration, inherited retinal degeneration, achromatopsia and cone diseases, and diabetic retinopathy. In some chapters, we highlight the complex interactions between the ER and other intracellular organelles focusing on mitochondria and illustrate how ER stress signaling regulates common cellular stress pathways such as autophagy. We also touch upon the integrated stress response in retinal degeneration and diabetic retinopathy. Finally, we provide an update on the current development of pharmacological agents targeting the UPR response and discuss some unresolved questions and knowledge gaps to be addressed by future research.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States.
| | - Josh J Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Christopher R Starr
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eun-Jin Lee
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Karen Sophia Park
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Assylbek Zhylkibayev
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andy Medina
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jonathan H Lin
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
8
|
Cachoux VML, Balakireva M, Gracia M, Bosveld F, López-Gay JM, Maugarny A, Gaugué I, di Pietro F, Rigaud SU, Noiret L, Guirao B, Bellaïche Y. Epithelial apoptotic pattern emerges from global and local regulation by cell apical area. Curr Biol 2023; 33:4807-4826.e6. [PMID: 37827152 PMCID: PMC10681125 DOI: 10.1016/j.cub.2023.09.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/07/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023]
Abstract
Geometry is a fundamental attribute of biological systems, and it underlies cell and tissue dynamics. Cell geometry controls cell-cycle progression and mitosis and thus modulates tissue development and homeostasis. In sharp contrast and despite the extensive characterization of the genetic mechanisms of caspase activation, we know little about whether and how cell geometry controls apoptosis commitment in developing tissues. Here, we combined multiscale time-lapse microscopy of developing Drosophila epithelium, quantitative characterization of cell behaviors, and genetic and mechanical perturbations to determine how apoptosis is controlled during epithelial tissue development. We found that early in cell lives and well before extrusion, apoptosis commitment is linked to two distinct geometric features: a small apical area compared with other cells within the tissue and a small relative apical area with respect to the immediate neighboring cells. We showed that these global and local geometric characteristics are sufficient to recapitulate the tissue-scale apoptotic pattern. Furthermore, we established that the coupling between these two geometric features and apoptotic cells is dependent on the Hippo/YAP and Notch pathways. Overall, by exploring the links between cell geometry and apoptosis commitment, our work provides important insights into the spatial regulation of cell death in tissues and improves our understanding of the mechanisms that control cell number and tissue size.
Collapse
Affiliation(s)
- Victoire M L Cachoux
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Maria Balakireva
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Mélanie Gracia
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Floris Bosveld
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Jesús M López-Gay
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Aude Maugarny
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Isabelle Gaugué
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Florencia di Pietro
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Stéphane U Rigaud
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Lorette Noiret
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Boris Guirao
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France.
| | - Yohanns Bellaïche
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France.
| |
Collapse
|
9
|
Melamed D, Choi A, Reilein A, Tavaré S, Kalderon D. Spatial regulation of Drosophila ovarian Follicle Stem Cell division rates and cell cycle transitions. PLoS Genet 2023; 19:e1010965. [PMID: 37747936 PMCID: PMC10553835 DOI: 10.1371/journal.pgen.1010965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/05/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023] Open
Abstract
Drosophila ovarian Follicle Stem Cells (FSCs) present a favorable paradigm for understanding how stem cell division and differentiation are balanced in communities where those activities are independent. FSCs also allow exploration of how this balance is integrated with spatial stem cell heterogeneity. Posterior FSCs become proliferative Follicle Cells (FCs), while anterior FSCs become quiescent Escort Cells (ECs) at about one fourth the frequency. A single stem cell can nevertheless produce both FCs and ECs because it can move between anterior and posterior locations. Studies based on EdU incorporation to approximate division rates suggested that posterior FSCs divide faster than anterior FSCs. However, direct measures of cell cycle times are required to ascertain whether FC output requires a net flow of FSCs from anterior to posterior. Here, by using live imaging and FUCCI cell-cycle reporters, we measured absolute division rates. We found that posterior FSCs cycle more than three times faster than anterior FSCs and produced sufficient new cells to match FC production. H2B-RFP dilution studies supported different cycling rates according to A/P location and facilitated live imaging, showing A/P exchange of FSCs in both directions, consistent with the dynamic equilibrium inferred from division rate measurements. Inversely graded Wnt and JAK-STAT pathway signals regulate FSC differentiation to ECs and FCs. JAK-STAT promotes both differentiation to FCs and FSC cycling, affording some coordination of these activities. When JAK-STAT signaling was manipulated to be spatially uniform, the ratio of posterior to anterior division rates was reduced but remained substantial, showing that graded JAK-STAT signaling only partly explains the graded cycling of FSCs. By using FUCCI markers, we found a prominent G2/M cycling restriction of posterior FSCs together with an A/P graded G1/S restriction, that JAK-STAT signaling promotes both G1/S and G2/M transitions, and that PI3 kinase signaling principally stimulates the G2/M transition.
Collapse
Affiliation(s)
- David Melamed
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
| | - Aaron Choi
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
| | - Amy Reilein
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
| | - Simon Tavaré
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
- Irving Institute for Cancer Dynamics & Department of Statistics, Columbia University, New York, New York State, United States of America
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
| |
Collapse
|
10
|
Sun X, Wang S, Miao X, Zeng S, Guo Y, Zhou A, Chen Y, Chen Y, Lv F, Fan Z, Wang Y, Xu Y, Li Z. TRIB1 regulates liver regeneration by antagonizing the NRF2-mediated antioxidant response. Cell Death Dis 2023; 14:372. [PMID: 37355685 PMCID: PMC10290656 DOI: 10.1038/s41419-023-05896-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
Robust regenerative response post liver injuries facilitates the architectural and functional recovery of the liver. Intrahepatic redox homeostasis plays a key role in liver regeneration. In the present study, we investigated the contributory role of Tribbles homolog 1 (Trib1), a pseudokinase, in liver regeneration and the underlying mechanism. We report that Trib1 expression was transiently down-regulated in animal and cell models of liver regeneration. Further analysis revealed that hepatocyte growth factor (HGF) repressed Trib1 transcription by evicting liver X receptor (LXRα) from the Trib1 promoter. Knockdown of Trib1 enhanced whereas over-expression of Trib1 suppressed liver regeneration after partial hepatectomy in mice. Of interest, regulation of liver regenerative response by Trib1 coincided with alterations of intracellular ROS levels, GSH levels, and antioxidant genes. Transcriptional assays suggested that Trib1 influenced cellular redox status by attenuating nuclear factor erythroid 2-related factor 2 (Nrf2) activity. Mechanistically, Trib1 interacted with the C-terminus of Nrf2 thus masking a potential nuclear localization signal (NLS) and blocking nuclear accumulation of Nrf2. Finally, correlation between Trib1 expression, Nrf2 nuclear localization, and cell proliferation was identified in liver specimens taken from patients with acute liver failure. In conclusion, our data unveil a novel pathway that depicts Trib1 as a critical link between intracellular redox homeostasis and cell proliferation in liver regeneration.
Collapse
Affiliation(s)
- Xinyue Sun
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Shuai Wang
- Department of General Surgery, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Xiulian Miao
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Sheng Zeng
- Stem Cell Center, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Yan Guo
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Anqi Zhou
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Ying Chen
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yifei Chen
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Fangqiao Lv
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhiwen Fan
- Department of Pathology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Yutong Wang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Yong Xu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China.
| | - Zilong Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China.
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China.
| |
Collapse
|
11
|
Osswald M, Barros-Carvalho A, Carmo AM, Loyer N, Gracio PC, Sunkel CE, Homem CCF, Januschke J, Morais-de-Sá E. aPKC regulates apical constriction to prevent tissue rupture in the Drosophila follicular epithelium. Curr Biol 2022; 32:4411-4427.e8. [PMID: 36113470 PMCID: PMC9632327 DOI: 10.1016/j.cub.2022.08.063] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/14/2022] [Accepted: 08/22/2022] [Indexed: 01/02/2023]
Abstract
Apical-basal polarity is an essential epithelial trait controlled by the evolutionarily conserved PAR-aPKC polarity network. Dysregulation of polarity proteins disrupts tissue organization during development and in disease, but the underlying mechanisms are unclear due to the broad implications of polarity loss. Here, we uncover how Drosophila aPKC maintains epithelial architecture by directly observing tissue disorganization after fast optogenetic inactivation in living adult flies and ovaries cultured ex vivo. We show that fast aPKC perturbation in the proliferative follicular epithelium produces large epithelial gaps that result from increased apical constriction, rather than loss of apical-basal polarity. Accordingly, we can modulate the incidence of epithelial gaps by increasing and decreasing actomyosin-driven contractility. We traced the origin of these large epithelial gaps to tissue rupture next to dividing cells. Live imaging shows that aPKC perturbation induces apical constriction in non-mitotic cells within minutes, producing pulling forces that ultimately detach dividing and neighboring cells. We further demonstrate that epithelial rupture requires a global increase of apical constriction, as it is prevented by the presence of non-constricting cells. Conversely, a global induction of apical tension through light-induced recruitment of RhoGEF2 to the apical side is sufficient to produce tissue rupture. Hence, our work reveals that the roles of aPKC in polarity and actomyosin regulation are separable and provides the first in vivo evidence that excessive tissue stress can break the epithelial barrier during proliferation.
Collapse
Affiliation(s)
- Mariana Osswald
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - André Barros-Carvalho
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana M Carmo
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Nicolas Loyer
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD5 1EH, UK
| | - Patricia C Gracio
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1150-199 Lisbon, Portugal
| | - Claudio E Sunkel
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Catarina C F Homem
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1150-199 Lisbon, Portugal
| | - Jens Januschke
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD5 1EH, UK
| | - Eurico Morais-de-Sá
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
12
|
Donker L, Houtekamer R, Vliem M, Sipieter F, Canever H, Gómez-González M, Bosch-Padrós M, Pannekoek WJ, Trepat X, Borghi N, Gloerich M. A mechanical G2 checkpoint controls epithelial cell division through E-cadherin-mediated regulation of Wee1-Cdk1. Cell Rep 2022; 41:111475. [PMID: 36223752 PMCID: PMC7617330 DOI: 10.1016/j.celrep.2022.111475] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 07/07/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
Epithelial cell divisions are coordinated with cell loss to preserve epithelial integrity. However, how epithelia adapt their rate of cell division to changes in cell number, for instance during homeostatic turnover or wounding, is not well understood. Here, we show that epithelial cells sense local cell density through mechanosensitive E-cadherin adhesions to control G2/M cell-cycle progression. As local cell density increases, tensile forces on E-cadherin adhesions are reduced, which prompts the accumulation of the G2 checkpoint kinase Wee1 and downstream inhibitory phosphorylation of Cdk1. Consequently, dense epithelia contain a pool of cells that are temporarily halted in G2 phase. These cells are readily triggered to divide following epithelial wounding due to the consequent increase in intercellular forces and resulting degradation of Wee1. Our data collectively show that epithelial cell division is controlled by a mechanical G2 checkpoint, which is regulated by cell-density-dependent intercellular forces sensed and transduced by E-cadherin adhesions.
Collapse
Affiliation(s)
- Lisa Donker
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 Utrecht, the Netherlands
| | - Ronja Houtekamer
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 Utrecht, the Netherlands
| | - Marjolein Vliem
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 Utrecht, the Netherlands
| | - François Sipieter
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Helena Canever
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Manuel Gómez-González
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Spain
| | - Miquel Bosch-Padrós
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Spain
| | - Willem-Jan Pannekoek
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 Utrecht, the Netherlands
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Spain; Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08028 Barcelona, Spain
| | - Nicolas Borghi
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Martijn Gloerich
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 Utrecht, the Netherlands.
| |
Collapse
|
13
|
Haraguchi-Suzuki K, Kawabata-Iwakawa R, Suzuki T, Suto T, Takazawa T, Saito S. Local anesthetic lidocaine-inducible gene, growth differentiation factor-15 suppresses the growth of cancer cell lines. Sci Rep 2022; 12:14520. [PMID: 36008442 PMCID: PMC9411556 DOI: 10.1038/s41598-022-18572-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022] Open
Abstract
Administration of local anesthetics, such as lidocaine, in the perioperative period improves outcomes of cancer patients. However, its precise mechanism is still unresolved. The growth of human cancer cell lines, including HeLa cells, are suppressed by lidocaine treatment. We identified that growth differentiation factor-15 (GDF-15) was commonly upregulated in lidocaine-treated cancer cell lines. GDF-15 is a divergent member of the transforming growth factor-β (TGF-β) superfamily and it is produced as an unprocessed pro-protein form and then cleaved to generate a mature form. In lidocaine-treated HeLa cells, increased production of GDF-15 in the endoplasmic reticulum (ER) was observed and unprocessed pro-protein form of GDF-15 was secreted extracellularly. Further, lidocaine induced apoptosis and apoptosis-inducible Tribbles homologue 3 (TRIB3) was also commonly upregulated in lidocaine-treated cancer cell lines. In addition, transcription factor C/EBP homologous protein (CHOP), which is a positive regulator of not only GDF-15 but TRIB3 was also induced by lidocaine. Lidocaine-induced growth suppression and apoptosis was suppressed by knockdown of GDF-15 or TRIB3 expression by small interference RNA (siRNA). These observations suggest that lidocaine suppresses the growth of cancer cells through increasing GDF-15 and TRIB3 expression, suggesting its potential application as cancer therapy.
Collapse
Affiliation(s)
- Keiko Haraguchi-Suzuki
- Intensive Care Unit, Gunma University Hospital, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Initiative for Advanced Research, Gunma University, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Toru Suzuki
- Laboratory for Immunogenetics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Takashi Suto
- Department of Anesthesiology, Gunma University Hospital, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tomonori Takazawa
- Intensive Care Unit, Gunma University Hospital, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Shigeru Saito
- Department of Anesthesiology, Gunma University Hospital, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
14
|
Yoshida T, Aonuma H, Otsuka S, Ichimura H, Saiki E, Hashimoto K, Ote M, Matsumoto S, Iwadate K, Miyawaki T, Kanuka H. A human tissue-based assay identifies a novel carrion blowfly strain for maggot debridement therapy. Sci Rep 2022; 12:12191. [PMID: 35842442 PMCID: PMC9288425 DOI: 10.1038/s41598-022-16253-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/07/2022] [Indexed: 11/15/2022] Open
Abstract
Maggot debridement therapy (MDT) is a form of therapeutic wound treatment in which live fly larvae are used intentionally to debride necrotic tissues. MDT has been widely used to treat chronic wounds in humans or animals, such as diabetic foot ulcers. Larvae of a carrion blowfly, Lucilia sericata (green bottle fly), debride wounds by consuming necrotic tissue and removing pathogenic bacteria, promoting effective wound healing. Most medical L. sericata strains were initially collected from natural environments using animal meat as bait and reared on artificial protein-rich media or ground meat. It remains to be examined which strain would be more appropriate for MDT, whereas any method for evaluating the fly’s therapeutic potential in humans has not been available. A feeding assay was developed using minced human tissues obtained from surgical waste. To establish L. sericata strains highly eligible for MDT, carrion fly larvae were collected from 45 corpses subjected to forensic autopsy (such as decomposed bodies). Four corpse-derived L. sericata strains were obtained and evaluated using the feeding assay. One strain showed that its feeding activity was 1.4 times higher than the control strain used in conventional MDT. The body length of the adult fly of the corpse-derived strain was longer than the control, which was consistent with the observation that its cell size was enlarged. The human tissue-based assay developed in this study accurately evaluated the ability of fly larvae to debride necrotic wounds. The L. sericata strain newly established from human corpses harboring high feeding activity may offer a clinically significant improvement in MDT.
Collapse
Affiliation(s)
- Takuma Yoshida
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Center for Medical Entomology, The Jikei University School of Medicine, Tokyo, Japan.,Department of Plastic and Reconstructive Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroka Aonuma
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Center for Medical Entomology, The Jikei University School of Medicine, Tokyo, Japan
| | - Saori Otsuka
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Center for Medical Entomology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hidetoshi Ichimura
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Center for Medical Entomology, The Jikei University School of Medicine, Tokyo, Japan
| | - Erisha Saiki
- Center for Medical Entomology, The Jikei University School of Medicine, Tokyo, Japan.,Laboratory Animal Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Kosei Hashimoto
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Center for Medical Entomology, The Jikei University School of Medicine, Tokyo, Japan
| | - Manabu Ote
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan.,Center for Medical Entomology, The Jikei University School of Medicine, Tokyo, Japan
| | - Sari Matsumoto
- Department of Forensic Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kimiharu Iwadate
- Department of Forensic Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takeshi Miyawaki
- Department of Plastic and Reconstructive Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Hirotaka Kanuka
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan. .,Center for Medical Entomology, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
15
|
Macabenta F, Sun HT, Stathopoulos A. BMP-gated cell-cycle progression drives anoikis during mesenchymal collective migration. Dev Cell 2022; 57:1683-1693.e3. [PMID: 35709766 PMCID: PMC9339487 DOI: 10.1016/j.devcel.2022.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/02/2022] [Accepted: 05/20/2022] [Indexed: 11/03/2022]
Abstract
Tissue homeostasis involves the elimination of abnormal cells to avoid compromised patterning and function. Although quality control through cell competition is well studied in epithelial tissues, it is unknown if and how homeostasis is regulated in mesenchymal collectives. Here, we demonstrate that collectively migrating Drosophila muscle precursors utilize both fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) signaling to promote homeostasis via anoikis, a form of cell death in response to substrate de-adhesion. Cell-cycle-regulated expression of the cell death gene head involution defective is responsible for caudal visceral mesoderm (CVM) anoikis. The secreted BMP ligand drives cell-cycle progression via a visceral mesoderm-specific cdc25/string enhancer to synchronize collective proliferation, as well as apoptosis of cells that have lost access to substrate-derived FGF. Perturbation of BMP-dependent cell-cycle progression is sufficient to confer anoikis resistance to mismigrating cells and thus facilitate invasion of other tissues. This BMP-gated cell-cycle checkpoint defines a quality control mechanism during mesenchymal collective migration.
Collapse
Affiliation(s)
- Frank Macabenta
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Hsuan-Te Sun
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Angelike Stathopoulos
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA.
| |
Collapse
|
16
|
Chen Q, Song Y, Yang N, Ai X, Pu L, Kong L. Aging deteriorated liver Ischemia and reperfusion injury by suppressing Tribble's proteins 1 mediated macrophage polarization. Bioengineered 2022; 13:14519-14533. [PMID: 36694470 PMCID: PMC9995131 DOI: 10.1080/21655979.2022.2090218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Aggravated liver injury has been reported in aged ischemia/reperfusion-stressed livers; however, the mechanism of aged macrophage inflammatory regulation is not well understood. Here, we found that the adaptor protein TRIB1 plays a critical role in the differentiation of macrophages and the inflammatory response in the liver after ischemia/reperfusion injury. In the present study, we determined that aging promoted macrophage-mediated liver injury and that inflammation was mainly responsible for lower M2 polarization in liver transplantation-exposed humans post I/R. Young and aged mice were subjected to hepatic I/R modeling and showed that aging aggravated liver injury and suppressed macrophage TRIB1 protein expression and anti-inflammatory function in I/R-stressed livers. Restoration of TRIB1 is mediated by lentiviral infection-induced macrophage anti-inflammatory M2 polarization and alleviated hepatic I/R injury. Moreover, TRIB1 overexpression in macrophages facilitates M2 polarization and anti-inflammation by activating MEK1-ERK1/2 signaling under IL-4 stimulation. Taken together, our results demonstrated that aging promoted hepatic I/R injury by suppressing TRIB1-mediated MEK1-induced macrophage M2 polarization and anti-inflammatory function.
Collapse
Affiliation(s)
- Qi Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Department of General Surgery, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yating Song
- Department of Bariatric and Metabolic Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ningli Yang
- Department of Bariatric and Metabolic Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoming Ai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lianbao Kong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
17
|
Danger R, Feseha Y, Brouard S. The Pseudokinase TRIB1 in Immune Cells and Associated Disorders. Cancers (Basel) 2022; 14:cancers14041011. [PMID: 35205759 PMCID: PMC8869936 DOI: 10.3390/cancers14041011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary TRIB1 is at the center of major cell signaling pathways. In this review, we describe its role in immune cells and highlight TRIB1 interacting partners which suggests cell-specific functions and that TRIB1 is involved in cellular homeostasis and also in different cancers and immune-related disorders. Abstract Research advances in Tribbles homolog (TRIB) genes have established the consensus that this protein family plays roles in diverse biological conditions and regulates intracellular signaling networks and several human diseases. In this review, we focus on one member of the family, TRIB1, and its role at the crossroads of immune signaling. TRIB1 directly interacts with transcription factors such as FOXP3 and C/EBPα, with several signaling molecules such as MEK1 and MALT1 and directly acts on key cell signaling pathways such as the MAPK and NF-κB pathways. Altogether, these interactions emphasize that TRIB1 is at the center of major cell signaling pathways while TRIB1 has cell-specific roles, potentially depending on the expressing cells and binding partners. In this review, we describe its roles in immune cells and highlight the interacting partners explaining these functions which suggests TRIB1 as a precise mediator of cellular homeostasis as well as in different cancers and immune-related disorders.
Collapse
Affiliation(s)
- Richard Danger
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, F-44000 Nantes, France; (R.D.); (Y.F.)
| | - Yodit Feseha
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, F-44000 Nantes, France; (R.D.); (Y.F.)
| | - Sophie Brouard
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, F-44000 Nantes, France; (R.D.); (Y.F.)
- LabEx IGO “Immunotherapy, Graft, Oncology”, F-44000 Nantes, France
- Correspondence: ; Tel.: +33-240-087-842
| |
Collapse
|
18
|
Monga J, Adrianto I, Rogers C, Gadgeel S, Chitale D, Alumkal JJ, Beltran H, Zoubeidi A, Ghosh J. Tribbles 2 pseudokinase confers enzalutamide resistance in prostate cancer by promoting lineage plasticity. J Biol Chem 2022; 298:101556. [PMID: 34973338 PMCID: PMC8800106 DOI: 10.1016/j.jbc.2021.101556] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 02/03/2023] Open
Abstract
Enzalutamide, a second-generation antiandrogen, is commonly prescribed for the therapy of advanced prostate cancer, but enzalutamide-resistant, lethal, or incurable disease invariably develops. To understand the molecular mechanism(s) behind enzalutamide resistance, here, we comprehensively analyzed a range of prostate tumors and clinically relevant models by gene expression array, immunohistochemistry, and Western blot, which revealed that enzalutamide-resistant prostate cancer cells and tumors overexpress the pseudokinase, Tribbles 2 (TRIB2). Inhibition of TRIB2 decreases the viability of enzalutamide-resistant prostate cancer cells, suggesting a critical role of TRIB2 in these cells. Moreover, the overexpression of TRIB2 confers resistance in prostate cancer cells to clinically relevant doses of enzalutamide, and this resistance is lost upon inhibition of TRIB2. Interestingly, we found that TRIB2 downregulates the luminal markers androgen receptor and cytokeratin 8 in prostate cancer cells but upregulates the neuronal transcription factor BRN2 (Brain-2) and the stemness factor SOX2 (SRY-box 2) to induce neuroendocrine characteristics. Finally, we show that inhibition of either TRIB2 or its downstream targets, BRN2 or SOX2, resensitizes resistant prostate cancer cells to enzalutamide. Thus, TRIB2 emerges as a potential new regulator of transdifferentiation that confers enzalutamide resistance in prostate cancer cells via a mechanism involving increased cellular plasticity and lineage switching.
Collapse
Affiliation(s)
- Jitender Monga
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, Michigan, USA
| | - Indra Adrianto
- Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Craig Rogers
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, Michigan, USA; Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA
| | - Shirish Gadgeel
- Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA
| | - Dhananjay Chitale
- Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA; Department of Pathology, Henry Ford Health System, Detroit, Michigan, USA
| | - Joshi J Alumkal
- Department of Internal Medicine, Univeristy of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Amina Zoubeidi
- Department of Urologic Sciences, University of British Columbia and The Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Jagadananda Ghosh
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, Michigan, USA; Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan, USA.
| |
Collapse
|
19
|
Hernández-Quiles M, Baak R, Borgman A, den Haan S, Sobrevals Alcaraz P, van Es R, Kiss-Toth E, Vos H, Kalkhoven E. Comprehensive Profiling of Mammalian Tribbles Interactomes Implicates TRIB3 in Gene Repression. Cancers (Basel) 2021; 13:6318. [PMID: 34944947 PMCID: PMC8699236 DOI: 10.3390/cancers13246318] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/30/2022] Open
Abstract
The three human Tribbles (TRIB) pseudokinases have been implicated in a plethora of signaling and metabolic processes linked to cancer initiation and progression and can potentially be used as biomarkers of disease and prognosis. While their modes of action reported so far center around protein-protein interactions, the comprehensive profiling of TRIB interactomes has not been reported yet. Here, we have developed a robust mass spectrometry (MS)-based proteomics approach to characterize Tribbles' interactomes and report a comprehensive assessment and comparison of the TRIB1, -2 and -3 interactomes, as well as domain-specific interactions for TRIB3. Interestingly, TRIB3, which is predominantly localized in the nucleus, interacts with multiple transcriptional regulators, including proteins involved in gene repression. Indeed, we found that TRIB3 repressed gene transcription when tethered to DNA in breast cancer cells. Taken together, our comprehensive proteomic assessment reveals previously unknown interacting partners and functions of Tribbles proteins that expand our understanding of this family of proteins. In addition, our findings show that MS-based proteomics provides a powerful tool to unravel novel pseudokinase biology.
Collapse
Affiliation(s)
- Miguel Hernández-Quiles
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.H.-Q.); (R.B.); (A.B.); (S.d.H.)
| | - Rosalie Baak
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.H.-Q.); (R.B.); (A.B.); (S.d.H.)
| | - Anouska Borgman
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.H.-Q.); (R.B.); (A.B.); (S.d.H.)
| | - Suzanne den Haan
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.H.-Q.); (R.B.); (A.B.); (S.d.H.)
| | - Paula Sobrevals Alcaraz
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (P.S.A.); (R.v.E.); (H.V.)
| | - Robert van Es
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (P.S.A.); (R.v.E.); (H.V.)
| | - Endre Kiss-Toth
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2TN, UK;
| | - Harmjan Vos
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (P.S.A.); (R.v.E.); (H.V.)
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (M.H.-Q.); (R.B.); (A.B.); (S.d.H.)
| |
Collapse
|
20
|
Quiroz-Figueroa K, Vitali C, Conlon DM, Millar JS, Tobias JW, Bauer RC, Hand NJ, Rader DJ. TRIB1 regulates LDL metabolism through CEBPα-mediated effects on the LDL receptor in hepatocytes. J Clin Invest 2021; 131:146775. [PMID: 34779419 DOI: 10.1172/jci146775] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 09/21/2021] [Indexed: 12/20/2022] Open
Abstract
Genetic variants near the TRIB1 gene are highly significantly associated with plasma lipid traits and coronary artery disease. While TRIB1 is likely causal of these associations, the molecular mechanisms are not well understood. Here we sought to investigate how TRIB1 influences low density lipoprotein cholesterol (LDL-C) levels in mice. Hepatocyte-specific deletion of Trib1 (Trib1Δhep) in mice increased plasma cholesterol and apoB and slowed the catabolism of LDL-apoB due to decreased levels of LDL receptor (LDLR) mRNA and protein. Simultaneous deletion of the transcription factor CCAAT/enhancer-binding protein alpha (CEBPα) with TRIB1 eliminated the effects of TRIB1 on hepatic LDLR regulation and LDL catabolism. Using RNA-seq, we found that activating transcription factor 3 (Atf3) was highly upregulated in the livers of Trib1Δhep but not Trib1Δhep CebpaΔhep mice. ATF3 has been shown to directly bind to the CEBPα protein, and to repress the expression of LDLR by binding its promoter. Blunting the increase of ATF3 in Trib1Δhep mice reduced the levels of plasma cholesterol and partially attenuated the effects on LDLR. Based on these data, we conclude that deletion of Trib1 leads to a posttranslational increase in CEBPα, which increases ATF3 levels, thereby contributing to the downregulation of LDLR and increased plasma LDL-C.
Collapse
Affiliation(s)
| | - Cecilia Vitali
- Division of Translational Medicine and Human Genetics, Department of Medicine
| | - Donna M Conlon
- Division of Translational Medicine and Human Genetics, Department of Medicine
| | - John S Millar
- Division of Translational Medicine and Human Genetics, Department of Medicine
| | | | - Robert C Bauer
- Division of Translational Medicine and Human Genetics, Department of Medicine
| | - Nicholas J Hand
- Department of Genetics.,Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Department of Medicine.,Department of Genetics.,Department of Pediatrics, and.,Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
The Pseudokinase TRIB3 Negatively Regulates the HER2 Receptor Pathway and Is a Biomarker of Good Prognosis in Luminal Breast Cancer. Cancers (Basel) 2021; 13:cancers13215307. [PMID: 34771470 PMCID: PMC8582533 DOI: 10.3390/cancers13215307] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Breast cancer is the most frequent type of cancer in women. More than 70% of these tumors belong to the so-called luminal subtype which has, in general, a good prognosis. However, a fraction of patients with luminal breast cancer progress to an advanced or metastatic disease, which remains a major clinical and social problem. Therefore, it is crucial to identify novel biomarkers that help to predict the progression of the disease and to develop more efficacious therapeutic approaches to fight advanced luminal breast cancer. In this work we found that the increased expression of the protein tribbles pseudokinase 3 (TRIB3) is associated with a good prognosis and a better response to therapy in luminal breast cancer patients. We also found that this effect is at least in part due to the ability of TRIB3 to inhibit the activity of the oncogene HER2. Abstract Background: Tribbles pseudokinase 3 (TRIB3) has been proposed to both promote and restrict cancer generation and progression. However, the precise mechanisms that determine this dual role of TRIB3 in cancer remain to be understood. In this study we aimed to investigate the role of TRIB3 in luminal breast cancer, the most frequent subtype of this malignancy. Methods: We genetically manipulated TRIB3 expression in a panel of luminal breast cancer cell lines and analyzed its impact on cell proliferation, and the phosphorylation, levels, or subcellular localization of TRIB3 and other protein regulators of key signaling pathways in luminal breast cancer. We also analyzed TRIB3 protein expression in samples from luminal breast cancer patients and performed bioinformatic analyses in public datasets. Results: TRIB3 enhanced the proliferation and AKT phosphorylation in luminal A (HER2-) but decreased them in luminal B (HER2+) breast cancer cell lines. TRIB3 negatively regulated the stability of HER2 in luminal B breast cancer cell lines. TRIB3 expression was associated with increased disease-free survival and a better response to therapy in luminal breast cancer patients. Conclusions: Our findings support the exploration of TRIB3 as a potential biomarker and therapeutic target in luminal breast cancer.
Collapse
|
22
|
Kohrman AQ, Kim-Yip RP, Posfai E. Imaging developmental cell cycles. Biophys J 2021; 120:4149-4161. [PMID: 33964274 PMCID: PMC8516676 DOI: 10.1016/j.bpj.2021.04.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/30/2021] [Indexed: 01/05/2023] Open
Abstract
The last decade has seen a major expansion in development of live biosensors, the tools needed to genetically encode them into model organisms, and the microscopic techniques used to visualize them. When combined, these offer us powerful tools with which to make fundamental discoveries about complex biological processes. In this review, we summarize the availability of biosensors to visualize an essential cellular process, the cell cycle, and the techniques for single-cell tracking and quantification of these reporters. We also highlight studies investigating the connection of cellular behavior to the cell cycle, particularly through live imaging, and anticipate exciting discoveries with the combination of these technologies in developmental contexts.
Collapse
Affiliation(s)
- Abraham Q Kohrman
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Rebecca P Kim-Yip
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, New Jersey.
| |
Collapse
|
23
|
Zhang X, Zhang B, Zhang C, Sun G, Sun X. Current Progress in Delineating the Roles of Pseudokinase TRIB1 in Controlling Human Diseases. J Cancer 2021; 12:6012-6020. [PMID: 34539875 PMCID: PMC8425202 DOI: 10.7150/jca.51627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
Tribbles homolog 1 (TRIB1) is a member of the tribbles family of pseudoprotein kinases and is widely expressed in numerous tissues, such as bone marrow, skeletal muscle, liver, heart, and adipose tissue. It is closely associated with acute myeloid leukemia, prostate cancer, and tumor drug resistance, and can interfere with the hematopoietic stem cell cycle, promote tumor cell proliferation, and inhibit apoptosis. Recent studies have shown that TRIB1 can regulate acute and chronic inflammation by affecting the secretion of inflammatory factors, which is closely related to the occurrence of hyperlipidemia and cardiovascular diseases. Given the important biological functions of TRIB1, the reviews published till now are not sufficiently comprehensive. Therefore, this paper reviews the progress in TRIB1 research aimed at exploring its roles in cancer, hyperlipidemia, and cardiovascular disease, and providing a theoretical basis for further studies on the biological roles of TRIB1.
Collapse
Affiliation(s)
- Xuelian Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Chenyang Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China.,Key Laboratory of efficacy evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| |
Collapse
|
24
|
Ruiz-Cantos M, Hutchison CE, Shoulders CC. Musings from the Tribbles Research and Innovation Network. Cancers (Basel) 2021; 13:cancers13184517. [PMID: 34572744 PMCID: PMC8467127 DOI: 10.3390/cancers13184517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 11/16/2022] Open
Abstract
This commentary integrates historical and modern findings that underpin our understanding of the cell-specific functions of the Tribbles (TRIB) proteins that bear on tumorigenesis. We touch on the initial discovery of roles played by mammalian TRIB proteins in a diverse range of cell-types and pathologies, for example, TRIB1 in regulatory T-cells, TRIB2 in acute myeloid leukaemia and TRIB3 in gliomas; the origins and diversity of TRIB1 transcripts; microRNA-mediated (miRNA) regulation of TRIB1 transcript decay and translation; the substantial conformational changes that ensue on binding of TRIB1 to the transcription factor C/EBPα; and the unique pocket formed by TRIB1 to sequester its C-terminal motif bearing a binding site for the E3 ubiquitin ligase COP1. Unashamedly, the narrative is relayed through the perspective of the Tribbles Research and Innovation Network, and its establishment, progress and future ambitions: the growth of TRIB and COP1 research to hasten discovery of their cell-specific contributions to health and obesity-related cancers.
Collapse
|
25
|
High fat diet induced abnormalities in metabolism, growth, behavior, and circadian clock in Drosophila melanogaster. Life Sci 2021; 281:119758. [PMID: 34175317 DOI: 10.1016/j.lfs.2021.119758] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 01/15/2023]
Abstract
AIMS The current lifestyle trend has made people vulnerable to diabetes and related diseases. Years of scientific research have not been able to yield a cure to the disease completely. The current study aims to investigate a link between high-fat diet mediated diabesity and circadian rhythm in the Drosophila model and inferences that might help in establishing a cure to the dreaded disease. MAIN METHODS Several experimental methods including phenotypical, histological, biochemical, molecular, and behavioral assays were used in the study to detect obesity, diabetes, and changes in the circadian clock in the fly model. KEY FINDINGS The larva and adults of Drosophila melanogaster exposed to high-fat diet (HFD) displayed excess deposition of fat as lipid droplets and micronuclei formation in the gut, fat body, and crop. Larva and adults of HFD showed behavioral defects. The higher amount of triglyceride, glucose, trehalose in the whole body of larva and adult fly confirmed obesity-induced hyperglycemia. The overexpression of insulin gene (Dilp2) and tribble (trbl) gene expression confirmed insulin resistance in HFD adults. We also observed elevated ROS level, developmental delay, altered metal level, growth defects, locomotory rhythms, sleep fragmentation, and expression of circadian genes (per, tim, and clock) in HFD larva and adults. Thus, HFD impairs the metabolism to produce obesity, insulin resistance, disruption of clock, and circadian clock related co-mordities in D. melanogaster. SIGNIFICANCE The circadian gene expression provides an innovative perspective to understand and find a new treatment for type-II diabetes and circadian anomalies.
Collapse
|
26
|
Mayoral-Varo V, Jiménez L, Link W. The Critical Role of TRIB2 in Cancer and Therapy Resistance. Cancers (Basel) 2021; 13:cancers13112701. [PMID: 34070799 PMCID: PMC8198994 DOI: 10.3390/cancers13112701] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The Tribbles proteins are members of CAMK Ser/Thr protein kinase family. They are evolutionary conserved pseudokinases found in most tissues of eukaryotic organisms. This ubiquitously expressed protein family is characterized by containing a catalytically deficient kinase domain which lacks amino acid residues required for the productive interaction with ATP and metal ions. Tribbles proteins exert their biological functions mainly through direct interaction with MAPKK and AKT proteins, therefore regulating important pathways involved in cell proliferation, apoptosis and differentiation. Due to the role of MAPKK and AKT signalling in the context of cancer development, Tribbles proteins have been recently considered as biomarkers of cancer progression. Furthermore, as the atypical pseudokinase domain retains a binding platform for substrates, Tribbles targeting provides an attractive opportunity for drug development. Abstract The Tribbles pseudokinases family consists of TRIB1, TRIB2, TRIB3 and STK40 and, although evolutionarily conserved, they have distinctive characteristics. Tribbles members are expressed in a context and cell compartment-dependent manner. For example, TRIB1 and TRIB2 have potent oncogenic activities in vertebrate cells. Since the identification of Tribbles proteins as modulators of multiple signalling pathways, recent studies have linked their expression with several pathologies, including cancer. Tribbles proteins act as protein adaptors involved in the ubiquitin-proteasome degradation system, as they bridge the gap between substrates and E3 ligases. Between TRIB family members, TRIB2 is the most ancestral member of the family. TRIB2 is involved in protein homeostasis regulation of C/EBPα, β-catenin and TCF4. On the other hand, TRIB2 interacts with MAPKK, AKT and NFkB proteins, involved in cell survival, proliferation and immune response. Here, we review the characteristic features of TRIB2 structure and signalling and its role in many cancer subtypes with an emphasis on TRIB2 function in therapy resistance in melanoma, leukemia and glioblastoma. The strong evidence between TRIB2 expression and chemoresistance provides an attractive opportunity for targeting TRIB2.
Collapse
|
27
|
Stefanovska B, André F, Fromigué O. Tribbles Pseudokinase 3 Regulation and Contribution to Cancer. Cancers (Basel) 2021; 13:cancers13081822. [PMID: 33920424 PMCID: PMC8070086 DOI: 10.3390/cancers13081822] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Accumulating evidence supports a key function for Tribbles proteins in oncogenesis, both in leukemia and solid tumors. However, the exact role of these proteins is hard to define since in a context-dependent manner they can function as both oncogenes and tumor suppressors. Their complex role arises from the capacity to interact with a wide range of target molecules thereby acting as molecular scaffolds and signaling regulators of multiple pathways. This review focuses on one particular Tribbles family member, namely, TRIB3, addressing its gene and protein expression, as well as its role in cancer development and progression. Abstract The first Tribbles protein was identified as critical for the coordination of morphogenesis in Drosophila melanogaster. Three mammalian homologs were subsequently identified, with a structure similar to classic serine/threonine kinases, but lacking crucial amino acids for the catalytic activity. Thereby, the very weak ATP affinity classifies TRIB proteins as pseudokinases. In this review, we provide an overview of the regulation of TRIB3 gene expression at both transcriptional and post-translational levels. Despite the absence of kinase activity, TRIB3 interferes with a broad range of cellular processes through protein–protein interactions. In fact, TRIB3 acts as an adaptor/scaffold protein for many other proteins such as kinase-dependent proteins, transcription factors, ubiquitin ligases, or even components of the spliceosome machinery. We then state the contribution of TRIB3 to cancer development, progression, and metastasis. TRIB3 dysregulation can be associated with good or bad prognosis. Indeed, as TRIB3 interacts with and regulates the activity of many key signaling components, it can act as a tumor-suppressor or oncogene in a context-dependent manner.
Collapse
Affiliation(s)
- Bojana Stefanovska
- Inserm, UMR981, F-94805 Villejuif, France; (B.S.); (F.A.)
- Gustave Roussy, F-94805 Villejuif, France
- Orsay, Université Paris Saclay, F-91400 Gif-sur-Yvette, France
| | - Fabrice André
- Inserm, UMR981, F-94805 Villejuif, France; (B.S.); (F.A.)
- Gustave Roussy, F-94805 Villejuif, France
- Orsay, Université Paris Saclay, F-91400 Gif-sur-Yvette, France
- Department of Medical Oncology, Gustave Roussy, F-94805 Villejuif, France
| | - Olivia Fromigué
- Inserm, UMR981, F-94805 Villejuif, France; (B.S.); (F.A.)
- Gustave Roussy, F-94805 Villejuif, France
- Orsay, Université Paris Saclay, F-91400 Gif-sur-Yvette, France
- Correspondence: ; Tel.: +33-142114211
| |
Collapse
|
28
|
Control of Cell Growth and Proliferation by the Tribbles Pseudokinase: Lessons from Drosophila. Cancers (Basel) 2021; 13:cancers13040883. [PMID: 33672471 PMCID: PMC7923445 DOI: 10.3390/cancers13040883] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Tribbles pseudokinases represent a sub-branch of the CAMK (Ca2+/calmodulin-dependent protein kinase) subfamily and are associated with disease-associated signaling pathways associated with various cancers, including melanoma, lung, liver, and acute leukemia. The ability of this class of molecules to regulate cell proliferation was first recognized in the model organism Drosophila and the fruit fly genetic model and continues to provide insight into the molecular mechanism by which this family of adapter molecules regulates both normal development and disease associated with corruption of their proper regulation and function. Abstract The Tribbles (Trib) family of pseudokinase proteins regulate cell growth, proliferation, and differentiation during normal development and in response to environmental stress. Mutations in human Trib isoforms (Trib1, 2, and 3) have been associated with metabolic disease and linked to leukemia and the formation of solid tumors, including melanomas, hepatomas, and lung cancers. Drosophila Tribbles (Trbl) was the first identified member of this sub-family of pseudokinases and shares a conserved structure and similar functions to bind and direct the degradation of key mediators of cell growth and proliferation. Common Trib targets include Akt kinase (also known as protein kinase B), C/EBP (CAAT/enhancer binding protein) transcription factors, and Cdc25 phosphatases, leading to the notion that Trib family members stand athwart multiple pathways modulating their growth-promoting activities. Recent work using the Drosophila model has provided important insights into novel facets of conserved Tribbles functions in stem cell quiescence, tissue regeneration, metabolism connected to insulin signaling, and tumor formation linked to the Hippo signaling pathway. Here we highlight some of these recent studies and discuss their implications for understanding the complex roles Tribs play in cancers and disease pathologies.
Collapse
|
29
|
Obesity-induced TRB3 negatively regulates Brown adipose tissue function in mice. Biochem Biophys Res Commun 2021; 547:29-35. [PMID: 33592376 DOI: 10.1016/j.bbrc.2021.01.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/28/2021] [Indexed: 12/26/2022]
Abstract
Brown adipose tissue (BAT) and stimulating adaptive thermogenesis have been implicated as anti-obese and anti-diabetic tissues due to their ability to dissipate energy as heat by the expression of UCP1. We have recently demonstrated that TRB3 impairs differentiation of brown preadipocytes via inhibiting insulin signaling. However, the roles of the protein in BAT function and thermogenesis in vivo have not yet been established. For this study we tested the hypothesis that TRB3 mediates obesity- and diabetes-induced impairments in BAT differentiation and function, and that inhibition of TRB3 improves BAT function. TRB3 expression was increased in BAT from high-fat fed mice and ob/ob mice, which was associated with decreased UCP1 expression. Incubation of brown adipocytes with palmitate increased TRB3 expression and decreased UCP1. Knockout of TRB3 in mice displayed higher UCP1 expression in BAT and cold resistance. Incubation of brown adipocytes with ER stressors increased TRB3 but decreased UCP1 and ER stress markers were elevated in BAT from high-fat fed mice and ob/ob mice. Finally, high-fat feeding in TRB3KO mice were protected from obesity-induced glucose intolerance and displayed cold resistance and higher expression of BAT-specific markers. These data demonstrate that high-fat feeding and obesity increase TRB3 in BAT, resulting in impaired tissue function.
Collapse
|
30
|
Warma A, Lussier JG, Ndiaye K. Tribbles Pseudokinase 2 (TRIB2) Regulates Expression of Binding Partners in Bovine Granulosa Cells. Int J Mol Sci 2021; 22:ijms22041533. [PMID: 33546420 PMCID: PMC7913596 DOI: 10.3390/ijms22041533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
Members of the Tribbles (TRIB) family of pseudokinases are critical components of intracellular signal transduction pathways in physiological and pathological processes. TRIBs, including TRIB2, have been previously shown as signaling mediators and scaffolding proteins regulating numerous cellular events such as proliferation, differentiation and cell death through protein stability and activity. However, the signaling network associated with TRIB2 and its binding partners in granulosa cells during ovarian follicular development is not fully defined. We previously reported that TRIB2 is differentially expressed in growing dominant follicles while downregulated in ovulatory follicles following the luteinizing hormone (LH) surge or human chorionic gonadotropin (hCG) injection. In the present study, we used the yeast two-hybrid screening system and in vitro coimmunoprecipitation assays to identify and confirm TRIB2 interactions in granulosa cells (GCs) of dominant ovarian follicles (DFs), which yielded individual candidate binding partners including calmodulin 1 (CALM1), inhibin subunit beta A (INHBA), inositol polyphosphate phosphatase-like 1 (INPPL1), 5'-nucleotidase ecto (NT5E), stearoyl-CoA desaturase (SCD), succinate dehydrogenase complex iron sulfur subunit B (SDHB) and Ras-associated protein 14 (RAB14). Further analyses showed that all TRIB2 binding partners are expressed in GCs of dominant follicles but are differentially regulated throughout the different stages of follicular development. CRISPR/Cas9-driven inhibition along with pQE-driven overexpression of TRIB2 showed that TRIB2 differently regulates expression of binding partners, which reveals the importance of TRIB2 in the control of gene expression linked to various biological processes such as proliferation, differentiation, cell migration, apoptosis, calcium signaling and metabolism. These data provide a larger view of potential TRIB2-regulated signal transduction pathways in GCs and provide strong evidence that TRIB2 may act as a regulator of target genes during ovarian follicular development.
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Lineage commitment is governed by instructive and stochastic signals, which drive both active induction of the lineage program and repression of alternative fates. Eosinophil lineage commitment is driven by the ordered interaction of transcription factors, supported by cytokine signals. This review summarizes key findings in the study of eosinophil lineage commitment and examines new data investigating the factors that regulate this process. RECENT FINDINGS Recent and past studies highlight how intrinsic and extrinsic signals modulate transcription factor network and lineage decisions. Early action of the transcription factors C/EBPα and GATA binding protein-1 along with C/EBPε supports lineage commitment and eosinophil differentiation. This process is regulated and enforced by the pseudokinase Trib1, a regulator of C/EBPα levels. The cytokines interleukin (IL)-5 and IL-33 also support early eosinophil development. However, current studies suggest that these cytokines are not specifically required for lineage commitment. SUMMARY Together, recent evidence suggests a model where early transcription factor activity drives expression of key eosinophil genes and cytokine receptors to prime lineage commitment. Understanding the factors and signals that control eosinophil lineage commitment may guide therapeutic development for eosinophil-mediated diseases and provide examples for fate choices in other lineages.
Collapse
|
32
|
Lemke S, Kale G, Urbansky S. Comparing gastrulation in flies: Links between cell biology and the evolution of embryonic morphogenesis. Mech Dev 2020. [DOI: 10.1016/j.mod.2020.103648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
33
|
Niespolo C, Johnston JM, Deshmukh SR, Satam S, Shologu Z, Villacanas O, Sudbery IM, Wilson HL, Kiss-Toth E. Tribbles-1 Expression and Its Function to Control Inflammatory Cytokines, Including Interleukin-8 Levels are Regulated by miRNAs in Macrophages and Prostate Cancer Cells. Front Immunol 2020; 11:574046. [PMID: 33329538 PMCID: PMC7728618 DOI: 10.3389/fimmu.2020.574046] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/27/2020] [Indexed: 12/21/2022] Open
Abstract
The pseudokinase TRIB1 controls cell function in a range of contexts, by regulating MAP kinase activation and mediating protein degradation via the COP1 ubiquitin ligase. TRIB1 regulates polarization of macrophages and dysregulated Trib1 expression in murine models has been shown to alter atherosclerosis burden and adipose homeostasis. Recently, TRIB1 has also been implicated in the pathogenesis of prostate cancer, where it is often overexpressed, even in the absence of genetic amplification. Well described TRIB1 effectors include MAP kinases and C/EBP transcription factors, both in immune cells and in carcinogenesis. However, the mechanisms that regulate TRIB1 itself remain elusive. Here, we show that the long and conserved 3’untranslated region (3’UTR) of TRIB1 is targeted by miRNAs in macrophage and prostate cancer models. By using a systematic in silico analysis, we identified multiple “high confidence” miRNAs potentially binding to the 3’UTR of TRIB1 and report that miR-101-3p and miR-132-3p are direct regulators of TRIB1 expression and function. Binding of miR-101-3p and miR-132-3p to the 3’UTR of TRIB1 mRNA leads to an increased transcription and secretion of interleukin-8. Our data demonstrate that modulation of TRIB1 by miRNAs alters the inflammatory profile of both human macrophages and prostate cancer cells.
Collapse
Affiliation(s)
- Chiara Niespolo
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Jessica M Johnston
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Sumeet R Deshmukh
- Department of Molecular Biology and Biotechnology, Sheffield Institute for Nucleic Acids, University of Sheffield, Sheffield, United Kingdom
| | - Swapna Satam
- Institute for Diabetes and Cancer IDC, Helmholtz Center, Munich, Germany
| | - Ziyanda Shologu
- Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | - Ian M Sudbery
- Department of Molecular Biology and Biotechnology, Sheffield Institute for Nucleic Acids, University of Sheffield, Sheffield, United Kingdom
| | - Heather L Wilson
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Endre Kiss-Toth
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
34
|
Yang J, Luan J, Shen Y, Chen B. Developments in the production of platelets from stem cells (Review). Mol Med Rep 2020; 23:7. [PMID: 33179095 PMCID: PMC7673345 DOI: 10.3892/mmr.2020.11645] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/13/2020] [Indexed: 01/01/2023] Open
Abstract
Platelets are small pieces of cytoplasm that have become detached from the cytoplasm of mature megakaryocytes (MKs) in the bone marrow. Platelets modulate vascular system integrity and serve important role, particularly in hemostasis. With the rapid development of clinical medicine, the demand for platelet transfusion as a life‑saving intervention increases continuously. Stem cell technology appears to be highly promising for transfusion medicine, and the generation of platelets from stem cells would be of great value in the clinical setting. Furthermore, several studies have been undertaken to investigate the potential of producing platelets from stem cells. Initial success has been achieved in terms of the yields and function of platelets generated from stem cells. However, the requirements of clinical practice remain unmet. The aim of the present review was to focus on several sources of stem cells and factors that induce MK differentiation. Updated information on current research into the genetic regulation of megakaryocytopoiesis and platelet generation was summarized. Additionally, advanced strategies of platelet generation were reviewed and the progress made in this field was discussed.
Collapse
Affiliation(s)
- Jie Yang
- Department of Hematology and Oncology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Jianfeng Luan
- Jinling Hospital Department of Blood Transfusion, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yanfei Shen
- Medical School, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Baoan Chen
- Department of Hematology and Oncology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
35
|
Hsu SJ, Stow EC, Simmons JR, Wallace HA, Lopez AM, Stroud S, Labrador M. Mutations in the insulator protein Suppressor of Hairy wing induce genome instability. Chromosoma 2020; 129:255-274. [PMID: 33140220 DOI: 10.1007/s00412-020-00743-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/14/2022]
Abstract
Insulator proteins orchestrate the three-dimensional organization of the genome. Insulators function by facilitating communications between regulatory sequences and gene promoters, allowing accurate gene transcription regulation during embryo development and cell differentiation. However, the role of insulator proteins beyond genome organization and transcription regulation remains unclear. Suppressor of Hairy wing [Su(Hw)] is a Drosophila insulator protein that plays an important function in female oogenesis. Here we find that su(Hw) has an unsuspected role in genome stability during cell differentiation. We show that su(Hw) mutant developing egg chambers have poorly formed microtubule organization centers (MTOCs) in the germarium and display mislocalization of the anterior/posterior axis specification factor gurken in later oogenesis stages. Additionally, eggshells from partially rescued su(Hw) mutant female germline exhibit dorsoventral patterning defects. These phenotypes are very similar to phenotypes found in the important class of spindle mutants or in piRNA pathway mutants in Drosophila, in which defects generally result from the failure of germ cells to repair DNA damage. Similarities between mutations in su(Hw) and spindle and piRNA mutants are further supported by an excess of DNA damage in nurse cells, and because Gurken localization defects are partially rescued by mutations in the ATR (mei-41) and Chk1 (grapes) DNA damage response genes. Finally, we also show that su(Hw) mutants produce an elevated number of chromosome breaks in dividing neuroblasts from larval brains. Together, these findings suggest that Su(Hw) is necessary for the maintenance of genome integrity during Drosophila development, in both germline and dividing somatic cells.
Collapse
Affiliation(s)
- Shih-Jui Hsu
- Department of Biochemistry and Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA
| | - Emily C Stow
- Department of Biochemistry and Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA
| | - James R Simmons
- Department of Biochemistry and Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA
| | - Heather A Wallace
- Department of Biochemistry and Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA
| | - Andrea Mancheno Lopez
- Department of Biochemistry and Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA
| | - Shannon Stroud
- Department of Biochemistry and Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA
| | - Mariano Labrador
- Department of Biochemistry and Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
36
|
Strong IJT, Lei X, Chen F, Yuan K, O’Farrell PH. Interphase-arrested Drosophila embryos activate zygotic gene expression and initiate mid-blastula transition events at a low nuclear-cytoplasmic ratio. PLoS Biol 2020; 18:e3000891. [PMID: 33090988 PMCID: PMC7608951 DOI: 10.1371/journal.pbio.3000891] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 11/03/2020] [Accepted: 09/14/2020] [Indexed: 11/18/2022] Open
Abstract
Externally deposited eggs begin development with an immense cytoplasm and a single overwhelmed nucleus. Rapid mitotic cycles restore normality as the ratio of nuclei to cytoplasm (N/C) increases. A threshold N/C has been widely proposed to activate zygotic genome transcription and onset of morphogenesis at the mid-blastula transition (MBT). To test whether a threshold N/C is required for these events, we blocked N/C increase by down-regulating cyclin/Cdk1 to arrest early cell cycles in Drosophila. Embryos that were arrested two cell cycles prior to the normal MBT activated widespread transcription of the zygotic genome including genes previously described as N/C dependent. Zygotic transcription of these genes largely retained features of their regulation in space and time. Furthermore, zygotically regulated post-MBT events such as cellularization and gastrulation movements occurred in these cell cycle-arrested embryos. These results are not compatible with models suggesting that these MBT events are directly coupled to N/C. Cyclin/Cdk1 activity normally declines in tight association with increasing N/C and is regulated by N/C. By experimentally promoting the decrease in cyclin/Cdk1, we uncoupled MBT from N/C increase, arguing that N/C-guided down-regulation of cyclin/Cdk1 is sufficient for genome activation and MBT.
Collapse
Affiliation(s)
- Isaac J. T. Strong
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
| | - Xiaoyun Lei
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Chen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Patrick H. O’Farrell
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
37
|
Perez-Vale KZ, Peifer M. Orchestrating morphogenesis: building the body plan by cell shape changes and movements. Development 2020; 147:dev191049. [PMID: 32917667 PMCID: PMC7502592 DOI: 10.1242/dev.191049] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
During embryonic development, a simple ball of cells re-shapes itself into the elaborate body plan of an animal. This requires dramatic cell shape changes and cell movements, powered by the contractile force generated by actin and myosin linked to the plasma membrane at cell-cell and cell-matrix junctions. Here, we review three morphogenetic events common to most animals: apical constriction, convergent extension and collective cell migration. Using the fruit fly Drosophila as an example, we discuss recent work that has revealed exciting new insights into the molecular mechanisms that allow cells to change shape and move without tearing tissues apart. We also point out parallel events at work in other animals, which suggest that the mechanisms underlying these morphogenetic processes are conserved.
Collapse
Affiliation(s)
- Kia Z Perez-Vale
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark Peifer
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
38
|
TRIB3 destabilizes tumor suppressor PPARα expression through ubiquitin-mediated proteasome degradation in acute myeloid leukemia. Life Sci 2020; 257:118021. [PMID: 32621919 DOI: 10.1016/j.lfs.2020.118021] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/17/2020] [Accepted: 06/24/2020] [Indexed: 12/23/2022]
Abstract
AIMS Tribbles homolog 3 (TRIB3) is emerging as a multifunctional oncoprotein associated with various cellular events in different tumors. However, the regulatory mechanism of TRIB3 in acute myeloid leukemia (AML) remains unknown. This study aims to investigate the molecular mechanisms and uncover the functions of TRIB3 in AML. METHODS Western blotting and quantitative real-time PCR were used to analyze the expression levels of TRIB3, peroxisome proliferator-activated receptor α (PPARα), apoptosis markers and autophagy markers in AML cells. Flow cytometry was used to assess cell apoptosis. The interaction of TRIB3 and PPARα was evaluated by immunofluorescence, coimmunoprecipitation, and in vivo ubiquitination assays. KEY FINDINGS We demonstrated that downregulating TRIB3 in leukemic cells effectively induced apoptosis and autophagy by regulating the degradation of PPARα. Mechanistically, TRIB3 interacted with PPARα and contributed to its destabilization by promoting its ubiquitination. When PPARα was activated by its specific agonist clofibrate, the apoptosis and autophagy of AML cells were significantly enhanced. These results were confirmed by rescue experiments. Blocking PPARα expression using the PPARα inhibitor GW6471 reversed the functional influence of TRIB3 on AML cells. SIGNIFICANCE The aim of this study is to provide evidence of the degradation of PPARα by TRIB3 via ubiquitin-dependent proteasomal degradation. This process meditates the progression of AML and prolongs the survival of leukemic cells. As a result, these data indicate that TRIB3 is a novel and promising therapeutic target for AML treatment.
Collapse
|
39
|
Abstract
Cancer is a genetic disease that involves the gradual accumulation of mutations. Human tumours are genetically unstable. However, the current knowledge about the origins and implications of genomic instability in this disease is limited. Understanding the biology of cancer requires the use of animal models. Here, we review relevant studies addressing the implications of genomic instability in cancer by using the fruit fly, Drosophila melanogaster, as a model system. We discuss how this invertebrate has helped us to expand the current knowledge about the mechanisms involved in genomic instability and how this hallmark of cancer influences disease progression.
Collapse
Affiliation(s)
- Stephan U Gerlach
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Héctor Herranz
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Sun J, Macabenta F, Akos Z, Stathopoulos A. Collective Migrations of Drosophila Embryonic Trunk and Caudal Mesoderm-Derived Muscle Precursor Cells. Genetics 2020; 215:297-322. [PMID: 32487692 PMCID: PMC7268997 DOI: 10.1534/genetics.120.303258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 04/17/2020] [Indexed: 01/06/2023] Open
Abstract
Mesoderm migration in the Drosophila embryo is a highly conserved, complex process that is required for the formation of specialized tissues and organs, including the somatic and visceral musculature. In this FlyBook chapter, we will compare and contrast the specification and migration of cells originating from the trunk and caudal mesoderm. Both cell types engage in collective migrations that enable cells to achieve new positions within developing embryos and form distinct tissues. To start, we will discuss specification and early morphogenetic movements of the presumptive mesoderm, then focus on the coordinate movements of the two subtypes trunk mesoderm and caudal visceral mesoderm, ending with a comparison of these processes including general insights gained through study.
Collapse
Affiliation(s)
- Jingjing Sun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Frank Macabenta
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Zsuzsa Akos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Angelike Stathopoulos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| |
Collapse
|
41
|
Liu B, Gregor I, Müller HA, Großhans J. Fluorescence fluctuation analysis reveals PpV dependent Cdc25 protein dynamics in living embryos. PLoS Genet 2020; 16:e1008735. [PMID: 32251417 PMCID: PMC7162543 DOI: 10.1371/journal.pgen.1008735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/16/2020] [Accepted: 03/25/2020] [Indexed: 12/21/2022] Open
Abstract
The protein phosphatase Cdc25 is a key regulator of the cell cycle by activating Cdk-cyclin complexes. Cdc25 is regulated by its expression levels and post-translational mechanisms. In early Drosophila embryogenesis, Cdc25/Twine drives the fast and synchronous nuclear cycles. A pause in the cell cycle and the remodeling to a more generic cell cycle mode with a gap phase are determined by Twine inactivation and destruction in early interphase 14, in response to zygotic genome activation. Although the pseudokinase Tribbles contributes to the timely degradation of Twine, Twine levels are controlled by additional yet unknown post-translational mechanisms. Here, we apply a non-invasive method based on fluorescence fluctuation analysis (FFA) to record the absolute concentration profiles of Twine with minute-scale resolution in single living embryos. Employing this assay, we found that Protein phosphatase V (PpV), the homologue of the catalytic subunit of human PP6, ensures appropriately low Twine protein levels at the onset of interphase 14. PpV controls directly or indirectly the phosphorylation of Twine at multiple serine and threonine residues as revealed by phosphosite mapping. Mutational analysis confirmed that these sites are involved in control of Twine protein dynamics, and cell cycle remodeling is delayed in a fraction of the phosphosite mutant embryos. Our data reveal a novel mechanism for control of Twine protein levels and their significance for embryonic cell cycle remodeling.
Collapse
Affiliation(s)
- Boyang Liu
- Fachbereich Biologie (FB17), Philipps-Universität Marburg, Marburg, Germany
- Institut für Entwicklungsbiochemie, Universitätsmedizin, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Ingo Gregor
- Drittes Physikalisches Institut, Georg-August-Universität Göttingen, Göttingen, Germany
| | - H.-Arno Müller
- Fachgebiet Entwicklungsgenetik, Institut für Biologie, Universität Kassel, Kassel, Germany
| | - Jörg Großhans
- Fachbereich Biologie (FB17), Philipps-Universität Marburg, Marburg, Germany
- Institut für Entwicklungsbiochemie, Universitätsmedizin, Georg-August-Universität Göttingen, Göttingen, Germany
| |
Collapse
|
42
|
Xie X, Yang X, Wu J, Ma J, Wei W, Fei X, Wang M. Trib1 Contributes to Recovery From Ischemia/Reperfusion-Induced Acute Kidney Injury by Regulating the Polarization of Renal Macrophages. Front Immunol 2020; 11:473. [PMID: 32265926 PMCID: PMC7098949 DOI: 10.3389/fimmu.2020.00473] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/28/2020] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence suggests that macrophage polarization is involved in the recovery from ischemia-reperfusion (I/R)-induced acute kidney injury (AKI), implying that the regulation of macrophage polarization homeostasis might mediate AKI recovery. Trib1 is a key regulator of macrophage differentiation, but its role in AKI remains unclear. Here, we aimed to investigate the role of Trib1 and its link with the macrophage phenotype in the process of adaptive recovery from I/R-induced renal injury. Lentiviral vector-mediated RNA interference (RNAi) was used to knock down Trib1 expression in vitro and in vivo, and a mouse model of moderate AKI was established by the induction of I/R injury. Renal function measurements and inflammatory factors were determined by the corresponding kits. Histomorphology was assessed by hematoxylin-eosin, Masson and PAS staining. Western blot and flow cytometry were employed for the analysis of signal transduction, cell apoptosis and macrophage phenotypes. Trib1 knockdown inhibited cell viability of tubular epithelial cells (TECs) by inhibiting proliferation and enhancing apoptosis in vitro. I/R-induced AKI significantly impaired renal function in mice via increasing the levels of BUN, Scr, NGAL and renal tubular damage, leading to renal fibrosis from days 1 to 3. Through the adaptive self-repair mechanism, renal dysfunction recovered over time and returned to almost normal levels on day 28 after I/R intervention. However, Trib1 depletion worsened renal damage on day 3 and blunted the adaptive repair process of the renal tissue. Mechanistically, Trib1 inhibition suppressed renal tubular cell proliferation under adaptive self-repair conditions by affecting the expression of the proliferation-related proteins cyclin D1, cyclin B, p21, and p27, the apoptosis-related proteins Bcl-2 and Bax, and the fibrosis-related proteins collagen I and III. Furthermore, the M1/M2 macrophage ratio increased in the first 3 days and decreased from day 7 to day 28, consistent with changes in the expression of inflammatory factors, including TNFα, IL-6, IL-12, IL-10, and IL-13. Trib1 inhibition blocked macrophage polarization during adaptive recovery from I/R-induced moderate AKI. Our results show that Trib1 plays a role in kidney recovery and regeneration via the regulation of renal tubular cell proliferation by affecting macrophage polarization. Thus, Trib1 might be a viable therapeutic target to improve renal adaptive repair following I/R injury.
Collapse
Affiliation(s)
- Xiangcheng Xie
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiu Yang
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junxia Wu
- Department of Nephrology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jilin Ma
- Division of Nephrology, Zhejiang Traditional Chinese Medicine and Western Medicine Hospital, Hangzhou, China
| | - Wei Wei
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Fei
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Wang
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
43
|
Warma A, Ndiaye K. Functional effects of Tribbles homolog 2 in bovine ovarian granulosa cells†. Biol Reprod 2020; 102:1177-1190. [PMID: 32159216 DOI: 10.1093/biolre/ioaa030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/12/2019] [Accepted: 03/07/2020] [Indexed: 12/19/2022] Open
Abstract
Tribbles homologs (TRIB) 1, 2, and 3 represent atypical members of the serine/threonine kinase superfamily. We previously identified TRIB2 as a differentially expressed gene in granulosa cells (GCs) of bovine preovulatory follicles. The current study aimed to further investigate TRIB2 regulation and study its function in the ovary. GCs were collected from follicles at different developmental stages: small antral follicles (SF), dominant follicles (DF) at day 5 of the estrous cycle, and hCG-induced ovulatory follicles (OFs). RT-qPCR analyses showed greater expression of TRIB2 in GC of DF as compared to OF and a significant downregulation of TRIB2 steady-state mRNA amounts by hCG/LH, starting at 6 h through 24 h post-hCG as compared to 0 h. Specific anti-TRIB2 polyclonal antibodies were generated and western blot analysis confirmed TRIB2 downregulation by hCG at the protein level. In vitro studies showed that FSH stimulates TRIB2 expression in GC. Inhibition of TRIB2 using CRISPR/Cas9 resulted in a significant increase in PCNA expression and an increase in steroidogenic enzyme CYP19A1 expression, while TRIB2 overexpression tended to decrease GC proliferation. TRIB2 inhibition also resulted in a decrease in transcription factors connective tissue growth factor (CTGF) and ankyrin repeat domain-containing protein 1 (ANKRD1) expression, while TRIB2 overexpression increased CTGF and ANKRD1. Additionally, western blot analyses showed reduction in ERK1/2 (MAPK3/1) and p38MAPK (MAPK14) phosphorylation levels following TRIB2 inhibition, while TRIB2 overexpression increased p-ERK1/2 and p-p38MAPK. These results provide evidence that TRIB2 modulates MAPK signaling in GC and that TRIB2 could act as a regulator of GC proliferation and function, which could affect steroidogenesis during follicular development.
Collapse
Affiliation(s)
- Aly Warma
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Centre de Recherche en Reproduction et Fertilité (CRRF), Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Kalidou Ndiaye
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Centre de Recherche en Reproduction et Fertilité (CRRF), Université de Montréal, St-Hyacinthe, Québec, Canada
| |
Collapse
|
44
|
Zhou S, Liu S, Lin C, Li Y, Ye L, Wu X, Jian Y, Dai Y, Ouyang Y, Zhao L, Liu M, Song L, Xi M. TRIB3 confers radiotherapy resistance in esophageal squamous cell carcinoma by stabilizing TAZ. Oncogene 2020; 39:3710-3725. [PMID: 32157210 DOI: 10.1038/s41388-020-1245-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 02/06/2023]
Abstract
Radioresistance becomes the major obstacle to reduce tumor recurrence and improve prognosis in the treatment of esophageal squamous cell carcinoma (ESCC). Thus new strategies for radioresistant ESCC are urgently needed. Herein, we reported that tribbles pseudokinase 3 (TRIB3) serves as a key regulator of radioresistance in ESCC. TRIB3 is overexpressed in ESCC tissues and cell lines. High expression of TRIB3 significantly correlates with poor radiotherapy response and prognosis in ESCC patients. Upregulation of TRIB3 in ESCC cells conferred radioresistance in vitro and in vivo by interacting with TAZ thus impeding β-TrCP-mediated TAZ ubiquitination and degradation. Conversely, silencing TRIB3 sensitized ESCC cells to ionizing radiation. More importantly, TRIB3 was significantly correlated with TAZ activation in ESCC biopsies, and patients with high expression of both TRIB3 and TAZ suffered the worst radiotherapy response and survival. Our study uncovers the critical mechanism of ESCC resistance to radiotherapy, and provides a new pharmacological opportunity for developing a mechanism-based strategy to eliminate radioresistant ESCC in clinical practice.
Collapse
Affiliation(s)
- Sha Zhou
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Shiliang Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Chuyong Lin
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yue Li
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Liping Ye
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Xianqiu Wu
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yunting Jian
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yuhu Dai
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Sun Yat-senUniversity, Guangzhou, 510080, China
| | - Ying Ouyang
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Lei Zhao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Mengzhong Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Libing Song
- Department of Experimental Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| | - Mian Xi
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| |
Collapse
|
45
|
Quiescent Neural Stem Cells for Brain Repair and Regeneration: Lessons from Model Systems. Trends Neurosci 2020; 43:213-226. [PMID: 32209453 DOI: 10.1016/j.tins.2020.02.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/26/2020] [Accepted: 02/05/2020] [Indexed: 12/29/2022]
Abstract
Neural stem cells (NSCs) are multipotent progenitors that are responsible for producing all of the neurons and macroglia in the nervous system. In adult mammals, NSCs reside predominantly in a mitotically dormant, quiescent state, but they can proliferate in response to environmental inputs such as feeding or exercise. It is hoped that quiescent NSCs could be activated therapeutically to contribute towards repair in humans. This will require an understanding of quiescent NSC heterogeneities and regulation during normal physiology and following brain injury. Non-mammalian vertebrates (zebrafish and salamanders) and invertebrates (Drosophila) offer insights into brain repair and quiescence regulation that are difficult to obtain using rodent models alone. We review conceptual progress from these various models, a first step towards harnessing quiescent NSCs for therapeutic purposes.
Collapse
|
46
|
Ko CS, Kalakuntla P, Martin AC. Apical Constriction Reversal upon Mitotic Entry Underlies Different Morphogenetic Outcomes of Cell Division. Mol Biol Cell 2020; 31:1663-1674. [PMID: 32129704 PMCID: PMC7521848 DOI: 10.1091/mbc.e19-12-0673] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
During development, coordinated cell shape changes and cell divisions sculpt tissues. While these individual cell behaviors have been extensively studied, how cell shape changes and cell divisions that occur concurrently in epithelia influence tissue shape is less understood. We addressed this question in two contexts of the early Drosophila embryo: premature cell division during mesoderm invagination, and native ectodermal cell divisions with ectopic activation of apical contractility. Using quantitative live-cell imaging, we demonstrated that mitotic entry reverses apical contractility by interfering with medioapical RhoA signaling. While premature mitotic entry inhibits mesoderm invagination, which relies on apical constriction, mitotic entry in an artificially contractile ectoderm induced ectopic tissue invaginations. Ectopic invaginations resulted from medioapical myosin loss in neighboring mitotic cells. This myosin loss enabled nonmitotic cells to apically constrict through mitotic cell stretching. Thus, the spatial pattern of mitotic entry can differentially regulate tissue shape through signal interference between apical contractility and mitosis.
Collapse
Affiliation(s)
- Clint S Ko
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Prateek Kalakuntla
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Adam C Martin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| |
Collapse
|
47
|
Martin AC. The Physical Mechanisms of Drosophila Gastrulation: Mesoderm and Endoderm Invagination. Genetics 2020; 214:543-560. [PMID: 32132154 PMCID: PMC7054018 DOI: 10.1534/genetics.119.301292] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/21/2019] [Indexed: 12/14/2022] Open
Abstract
A critical juncture in early development is the partitioning of cells that will adopt different fates into three germ layers: the ectoderm, the mesoderm, and the endoderm. This step is achieved through the internalization of specified cells from the outermost surface layer, through a process called gastrulation. In Drosophila, gastrulation is achieved through cell shape changes (i.e., apical constriction) that change tissue curvature and lead to the folding of a surface epithelium. Folding of embryonic tissue results in mesoderm and endoderm invagination, not as individual cells, but as collective tissue units. The tractability of Drosophila as a model system is best exemplified by how much we know about Drosophila gastrulation, from the signals that pattern the embryo to the molecular components that generate force, and how these components are organized to promote cell and tissue shape changes. For mesoderm invagination, graded signaling by the morphogen, Spätzle, sets up a gradient in transcriptional activity that leads to the expression of a secreted ligand (Folded gastrulation) and a transmembrane protein (T48). Together with the GPCR Mist, which is expressed in the mesoderm, and the GPCR Smog, which is expressed uniformly, these signals activate heterotrimeric G-protein and small Rho-family G-protein signaling to promote apical contractility and changes in cell and tissue shape. A notable feature of this signaling pathway is its intricate organization in both space and time. At the cellular level, signaling components and the cytoskeleton exhibit striking polarity, not only along the apical-basal cell axis, but also within the apical domain. Furthermore, gene expression controls a highly choreographed chain of events, the dynamics of which are critical for primordium invagination; it does not simply throw the cytoskeletal "on" switch. Finally, studies of Drosophila gastrulation have provided insight into how global tissue mechanics and movements are intertwined as multiple tissues simultaneously change shape. Overall, these studies have contributed to the view that cells respond to forces that propagate over great distances, demonstrating that cellular decisions, and, ultimately, tissue shape changes, proceed by integrating cues across an entire embryo.
Collapse
Affiliation(s)
- Adam C Martin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
| |
Collapse
|
48
|
Abstract
Genome-wide association studies (GWAS) have identified hundreds of genomic loci in humans that are significantly associated with plasma cholesterol, triglycerides, and coronary artery disease. Although some loci contain genes with known regulatory roles in lipid metabolism and atherosclerosis, the majority were being implicated for the first time. The 8q24 locus, containing the gene TRIB1 ( Tribbles-1), is the only novel GWAS locus that associates with all 4 plasma lipid traits and coronary artery disease, an observation that has spurred immense interest in this locus. Subsequent in vivo loss and gain of function studies confirmed that Trib1 plays a role in hepatic lipid metabolism, validating the initial genetic observation. Yet, many challenges remain in discerning the nature of the association between the TRIB1 locus and cardiometabolic phenotypes. Is TRIB1 the causal gene at the 8q24 locus and what is the functional consequence of the associated noncoding variation? Is the relationship between TRIB1 and the transcription factor C/EBPα (CCAAT/enhancer-binding protein alpha) the primary molecular mechanism governing the genetic association or could it be an as yet unknown function for this interesting pseudokinase? Is hepatic TRIB1 the sole regulator of lipid metabolism or could extrahepatic TRIB1 play a role as well? The following review summarizes key findings related to these questions and highlights both the challenges and excitement in pursuing translational research of a novel gene in the post-GWAS era.
Collapse
Affiliation(s)
- Kavita S Jadhav
- From the Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York
| | - Robert C Bauer
- From the Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York
| |
Collapse
|
49
|
Gou J, Stotsky JA, Othmer HG. Growth control in the Drosophila wing disk. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1478. [PMID: 31917525 DOI: 10.1002/wsbm.1478] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
The regulation of size and shape is a fundamental requirement of biological development and has been a subject of scientific study for centuries, but we still lack an understanding of how organisms know when to stop growing. Imaginal wing disks of the fruit fly Drosophila melanogaster, which are precursors of the adult wings, are an archetypal tissue for studying growth control. The growth of the disks is dependent on many inter- and intra-organ factors such as morphogens, mechanical forces, nutrient levels, and hormones that influence gene expression and cell growth. Extracellular signals are transduced into gene-control signals via complex signal transduction networks, and since cells typically receive many different signals, a mechanism for integrating the signals is needed. Our understanding of the effect of morphogens on tissue-level growth regulation via individual pathways has increased significantly in the last half century, but our understanding of how multiple biochemical and mechanical signals are integrated to determine whether or not a cell decides to divide is still rudimentary. Numerous fundamental questions are involved in understanding the decision-making process, and here we review the major biochemical and mechanical pathways involved in disk development with a view toward providing a basis for beginning to understand how multiple signals can be integrated at the cell level, and how this translates into growth control at the level of the imaginal disk. This article is categorized under: Analytical and Computational Methods > Computational Methods Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Jia Gou
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Jay A Stotsky
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
50
|
Epithelial-Mesenchymal Plasticity in Circulating Tumor Cells, the Precursors of Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:11-34. [PMID: 32304077 DOI: 10.1007/978-3-030-35805-1_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Circulating tumor cells offer an unprecedented window into the metastatic cascade, and to some extent can be considered as intermediates in the process of metastasis. They exhibit dynamic oscillations in epithelial to mesenchymal plasticity and provide important opportunities for prognosis, therapy response monitoring, and targeting of metastatic disease. In this manuscript, we review the involvement of epithelial-mesenchymal plasticity in the early steps of metastasis and what we have learned about its contribution to genomic instability and genetic diversity, tumor progression and therapeutic responses using cell culture, mouse models and circulating tumor cells enriched from patients.
Collapse
|