1
|
Azlan A, Zhu L, Fukunaga R. Female-germline specific protein Sakura interacts with Otu and is crucial for germline stem cell renewal and differentiation and oogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616675. [PMID: 39651236 PMCID: PMC11623502 DOI: 10.1101/2024.10.04.616675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
During oogenesis, self-renewal and differentiation of germline stem cells (GSCs) must be tightly regulated. The Drosophila female germline serves as an excellent model for studying these regulatory mechanisms. Here, we report that a previously uncharacterized gene CG14545 , which we named sakura , is essential for oogenesis and female fertility in Drosophila . Sakura is predominantly expressed in the ovaries, particularly in the germline cells, including GSCs. sakura null mutant female flies display rudimentary ovaries with germline-less and tumorous phenotypes, fail to produce eggs, and are completely sterile. The germline-specific depletion of sakura impairs Dpp/BMP signaling, leading to aberrant bag-of-marbles ( bam ) expression, resulting in faulty differentiation and loss of GSCs. Additionally, sakura is necessary for normal piwi-interacting RNAs (piRNAs) levels and for proper localization of Ool8 RNA-binding protein (Orb) in developing oocytes. We identified Ovarian Tumor (Otu) as protein binding partner of Sakura, and we found that loss of otu phenocopies loss of sakura in ovaries. Thus, we identified Sakura as a crucial factor for GSC renewal and differentiation and oogenesis, and propose that Sakura and Otu function together in these processes.
Collapse
|
2
|
Huang YT, Hesting LL, Calvi BR. An unscheduled switch to endocycles induces a reversible senescent arrest that impairs growth of the Drosophila wing disc. PLoS Genet 2024; 20:e1011387. [PMID: 39226333 PMCID: PMC11398662 DOI: 10.1371/journal.pgen.1011387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/13/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogaster wing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| | - Lauren L Hesting
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| | - Brian R Calvi
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
3
|
Kulkarni M, Selarka K, Shravage BV. Monitoring Autophagy During Drosophila Oogenesis. Methods Mol Biol 2024. [PMID: 39120738 DOI: 10.1007/7651_2024_563] [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: 08/10/2024]
Abstract
Macroautophagy (autophagy hereafter) is an evolutionarily conserved mechanism that maintains the health of cells by degrading toxic proteins and damaged organelles within the lysosomes. Tissues like ovary are made up of heterogeneous cell types and each cell type has distinct levels of autophagy. Studying autophagy in a cell-type specific manner helps better understand the role of autophagy during oogenesis. Here, we describe assays for monitoring autophagy during oogenesis in Drosophila using the two protein markers, Atg8a and Ref(2)P.
Collapse
Affiliation(s)
- Mrunmayee Kulkarni
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, Maharashtra, India
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind, Pune, Maharashtra, India
| | - Karan Selarka
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, Maharashtra, India
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind, Pune, Maharashtra, India
| | - Bhupendra V Shravage
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, Maharashtra, India.
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind, Pune, Maharashtra, India.
- Department of Zoology, Savitribai Phule Pune University, Ganeshkhind, Pune, Maharashtra, India.
| |
Collapse
|
4
|
Vidaurre V, Song A, Li T, Ku WL, Zhao K, Qian J, Chen X. The Drosophila histone methyltransferase SET1 coordinates multiple signaling pathways in regulating male germline stem cell maintenance and differentiation. Development 2024; 151:dev202729. [PMID: 39007366 PMCID: PMC11369688 DOI: 10.1242/dev.202729] [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: 01/25/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Many tissue-specific adult stem cell lineages maintain a balance between proliferation and differentiation. Here, we study how the H3K4me3 methyltransferase Set1 regulates early-stage male germ cells in Drosophila. Early-stage germline-specific knockdown of Set1 results in temporally progressive defects, arising as germ cell loss and developing into overpopulated early-stage germ cells. These germline defects also impact the niche architecture and cyst stem cell lineage non-cell-autonomously. Additionally, wild-type Set1, but not the catalytically inactive Set1, rescues the Set1 knockdown phenotypes, highlighting the functional importance of the methyltransferase activity of Set1. Further, RNA-sequencing experiments reveal key signaling pathway components, such as the JAK-STAT pathway gene Stat92E and the BMP pathway gene Mad, which are upregulated upon Set1 knockdown. Genetic interaction assays support the functional relationships between Set1 and JAK-STAT or BMP pathways, as both Stat92E and Mad mutations suppress the Set1 knockdown phenotypes. These findings enhance our understanding of the balance between proliferation and differentiation in an adult stem cell lineage. The phenotype of germ cell loss followed by over-proliferation when inhibiting a histone methyltransferase also raises concerns about using their inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Velinda Vidaurre
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Annabelle Song
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Taibo Li
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wai Lim Ku
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20814, USA
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20814, USA
| | - Jiang Qian
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xin Chen
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD 20815, USA
| |
Collapse
|
5
|
Zhang R, Shi P, Xu S, Ming Z, Liu Z, He Y, Dai J, Matunis E, Xu J, Ma Q. Soma-germline communication drives sex maintenance in the Drosophila testis. Natl Sci Rev 2024; 11:nwae215. [PMID: 39183747 PMCID: PMC11342250 DOI: 10.1093/nsr/nwae215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 08/27/2024] Open
Abstract
In adult gonads, disruption of somatic sexual identity leads to defective gametogenesis and infertility. However, the underlying mechanisms by which somatic signals regulate germline cells to achieve proper gametogenesis remain unclear. In our previous study, we introduced the chinmoSex Transformation (chinmoST ) mutant Drosophila testis phenotype as a valuable model for investigating the mechanisms underlying sex maintenance. In chinmoST testes, depletion of the Janus Kinase-Signal Transducer and Activator of Transcription downstream effector Chinmo from somatic cyst stem cells (CySCs) feminizes somatic cyst cells and arrests germline differentiation. Here, we use single-cell RNA sequencing to uncover chinmoST -specific cell populations and their transcriptomic changes during sex transformation. Comparative analysis of intercellular communication networks between wild-type and chinmoST testes revealed disruptions in several soma-germline signaling pathways in chinmoST testes. Notably, the insulin signaling pathway exhibited significant enhancement in germline stem cells (GSCs). Chinmo cleavage under targets and tagmentation (CUT&Tag) assay revealed that Chinmo directly regulates two male sex determination factors, doublesex (dsx) and fruitless (fru), as well as Ecdysone-inducible gene L2 (ImpL2), a negative regulator of the insulin signaling pathway. Further genetic manipulations confirmed that the impaired gametogenesis observed in chinmoST testes was partly contributed by dysregulation of the insulin signaling pathway. In summary, our study demonstrates that somatic sex maintenance promotes normal spermatogenesis through Chinmo-mediated conserved sex determination and the insulin signaling pathway. Our work offers new insights into the complex mechanisms of somatic stem cell sex maintenance and soma-germline communication at the single-cell level. Additionally, our discoveries highlight the potential significance of stem cell sex instability as a novel mechanism contributing to testicular tumorigenesis.
Collapse
Affiliation(s)
- Rui Zhang
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Peiyu Shi
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Shuyang Xu
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhe Ming
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zicong Liu
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuanyuan He
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Junbiao Dai
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Erika Matunis
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jin Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Qing Ma
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
6
|
Stewart RK, Nguyen P, Laederach A, Volkan PC, Sawyer JK, Fox DT. Orb2 enables rare-codon-enriched mRNA expression during Drosophila neuron differentiation. Nat Commun 2024; 15:5270. [PMID: 38902233 PMCID: PMC11190236 DOI: 10.1038/s41467-024-48344-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 04/25/2024] [Indexed: 06/22/2024] Open
Abstract
Regulation of codon optimality is an increasingly appreciated layer of cell- and tissue-specific protein expression control. Here, we use codon-modified reporters to show that differentiation of Drosophila neural stem cells into neurons enables protein expression from rare-codon-enriched genes. From a candidate screen, we identify the cytoplasmic polyadenylation element binding (CPEB) protein Orb2 as a positive regulator of rare-codon-dependent mRNA stability in neurons. Using RNA sequencing, we reveal that Orb2-upregulated mRNAs in the brain with abundant Orb2 binding sites have a rare-codon bias. From these Orb2-regulated mRNAs, we demonstrate that rare-codon enrichment is important for mRNA stability and social behavior function of the metabotropic glutamate receptor (mGluR). Our findings reveal a molecular mechanism by which neural stem cell differentiation shifts genetic code regulation to enable critical mRNA stability and protein expression.
Collapse
Affiliation(s)
- Rebeccah K Stewart
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, USA
- Duke Regeneration Center, Duke University, Durham, NC, USA
| | - Patrick Nguyen
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, USA
| | - Alain Laederach
- Department of Biology, University of North Carolina, Chapel Hill, NC, USA
| | | | - Jessica K Sawyer
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, USA
- Duke Regeneration Center, Duke University, Durham, NC, USA
| | - Donald T Fox
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, USA.
- Duke Regeneration Center, Duke University, Durham, NC, USA.
| |
Collapse
|
7
|
Huang YT, Hesting LL, Calvi BR. An unscheduled switch to endocycles induces a reversible senescent arrest that impairs growth of the Drosophila wing disc. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.585098. [PMID: 38559130 PMCID: PMC10980049 DOI: 10.1101/2024.03.14.585098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogasterwing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| | - Lauren L. Hesting
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| | - Brian R. Calvi
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| |
Collapse
|
8
|
Chen J, Li C, Sheng Y, Zhang J, Pang L, Dong Z, Wu Z, Lu Y, Liu Z, Zhang Q, Guan X, Chen X, Huang J. Communication between the stem cell niche and an adjacent differentiation niche through miRNA and EGFR signaling orchestrates exit from the stem cell state in the Drosophila ovary. PLoS Biol 2024; 22:e3002515. [PMID: 38512963 PMCID: PMC10986965 DOI: 10.1371/journal.pbio.3002515] [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: 07/07/2023] [Revised: 04/02/2024] [Accepted: 01/22/2024] [Indexed: 03/23/2024] Open
Abstract
The signaling environment, or niche, often governs the initial difference in behavior of an adult stem cell and a derivative that initiates a path towards differentiation. The transition between an instructive stem cell niche and differentiation niche must generally have single-cell resolution, suggesting that multiple mechanisms might be necessary to sharpen the transition. Here, we examined the Drosophila ovary and found that Cap cells, which are key constituents of the germline stem cell (GSC) niche, express a conserved microRNA (miR-124). Surprisingly, loss of miR-124 activity in Cap cells leads to a defect in differentiation of GSC derivatives. We present evidence that the direct functional target of miR-124 in Cap cells is the epidermal growth factor receptor (EGFR) and that failure to limit EGFR expression leads to the ectopic expression of a key anti-differentiation BMP signal in neighboring somatic escort cells (ECs), which constitute a differentiation niche. We further found that Notch signaling connects EFGR activity in Cap cells to BMP expression in ECs. We deduce that the stem cell niche communicates with the differentiation niche through a mechanism that begins with the selective expression of a specific microRNA and culminates in the suppression of the major anti-differentiation signal in neighboring cells, with the functionally important overall role of sharpening the spatial distinction between self-renewal and differentiation environments.
Collapse
Affiliation(s)
- Jiani Chen
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, Institute of Crop Science, Plant Precision Breeding Academy, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Chaosqun Li
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Yifeng Sheng
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Junwei Zhang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Lan Pang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, Institute of Crop Science, Plant Precision Breeding Academy, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Zhi Dong
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Zhiwei Wu
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Yueqi Lu
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Zhiguo Liu
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Qichao Zhang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, Institute of Crop Science, Plant Precision Breeding Academy, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Xueying Guan
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, Institute of Crop Science, Plant Precision Breeding Academy, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Sanya, China
| | - Xuexin Chen
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Sanya, China
| | - Jianhua Huang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Vidaurre V, Song A, Li T, Ku WL, Zhao K, Qian J, Chen X. The Drosophila histone methyl-transferase SET1 coordinates multiple signaling pathways in regulating male germline stem cell maintenance and differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580277. [PMID: 38405894 PMCID: PMC10888844 DOI: 10.1101/2024.02.14.580277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Many cell types come from tissue-specific adult stem cells that maintain the balance between proliferation and differentiation. Here, we study how the H3K4me3 methyltransferase, Set1, regulates early-stage male germ cell proliferation and differentiation in Drosophila. Early-stage germline-specific knockdown of set1 results in a temporally progressed defects, arising as germ cell loss and developing to overpopulated early-stage germ cells. These germline defects also impact the niche architecture and cyst stem cell lineage in a non-cell-autonomous manner. Additionally, wild-type Set1, but not the catalytically inactive Set1, could rescue the set1 knockdown phenotypes, highlighting the functional importance of the methyl-transferase activity of the Set1 enzyme. Further, RNA-seq experiments reveal key signaling pathway components, such as the JAK-STAT pathway gene stat92E and the BMP pathway gene mad, that are upregulated upon set1 knockdown. Genetic interaction assays support the functional relationships between set1 and JAK-STAT or BMP pathways, as mutations of both the stat92E and mad genes suppress the set1 knockdown phenotypes. These findings enhance our understanding of the balance between proliferation and differentiation in an adult stem cell lineage. The germ cell loss followed by over-proliferation phenotypes when inhibiting a histone methyl-transferase raise concerns about using their inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Velinda Vidaurre
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Annabelle Song
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Taibo Li
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Wai Lim Ku
- Systems Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Keji Zhao
- Systems Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Jiang Qian
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Xin Chen
- Howard Hughes Medical Institute, Baltimore, Maryland, United States of America
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
10
|
Kakino K, Mon H, Ebihara T, Hino M, Masuda A, Lee JM, Kusakabe T. Comprehensive Transcriptome Analysis in the Testis of the Silkworm, Bombyx mori. INSECTS 2023; 14:684. [PMID: 37623394 PMCID: PMC10455414 DOI: 10.3390/insects14080684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023]
Abstract
Spermatogenesis is an important process in reproduction and is conserved across species, but in Bombyx mori, it shows peculiarities, such as the maintenance of spermatogonia by apical cells and fertilization by dimorphic spermatozoa. In this study, we attempted to characterize the genes expressed in the testis of B. mori, focusing on aspects of expression patterns and gene function by transcriptome comparisons between different tissues, internal testis regions, and Drosophila melanogaster. The transcriptome analysis of 12 tissues of B. mori, including those of testis, revealed the widespread gene expression of 20,962 genes and 1705 testis-specific genes. A comparative analysis of the stem region (SR) and differentiated regions (DR) of the testis revealed 4554 and 3980 specific-enriched genes, respectively. In addition, comparisons with D. melanogaster testis transcriptome revealed homologs of 1204 SR and 389 DR specific-enriched genes that were similarly expressed in equivalent regions of Drosophila testis. Moreover, gene ontology (GO) enrichment analysis was performed for SR-specific enriched genes and DR-specific enriched genes, and the GO terms of several biological processes were enriched, confirming previous findings. This study advances our understanding of spermatogenesis in B. mori and provides an important basis for future research, filling a knowledge gap between fly and mammalian studies.
Collapse
Affiliation(s)
- Kohei Kakino
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan; (K.K.); (H.M.); (T.E.)
| | - Hiroaki Mon
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan; (K.K.); (H.M.); (T.E.)
| | - Takeru Ebihara
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan; (K.K.); (H.M.); (T.E.)
| | - Masato Hino
- Laboratory of Sanitary Entomology, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan;
| | - Akitsu Masuda
- Laboratory of Creative Science for Insect Industries, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan; (A.M.); (J.M.L.)
| | - Jae Man Lee
- Laboratory of Creative Science for Insect Industries, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan; (A.M.); (J.M.L.)
| | - Takahiro Kusakabe
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan; (K.K.); (H.M.); (T.E.)
| |
Collapse
|
11
|
Stewart RK, Nguyen P, Laederach A, Volkan PC, Sawyer JK, Fox DT. Orb2 enables rare-codon-enriched mRNA expression during Drosophila neuron differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550700. [PMID: 37546801 PMCID: PMC10402044 DOI: 10.1101/2023.07.26.550700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Regulation of codon optimality is an increasingly appreciated layer of cell- and tissue-specific protein expression control. Here, we use codon-modified reporters to show that differentiation of Drosophila neural stem cells into neurons enables protein expression from rare-codon-enriched genes. From a candidate screen, we identify the cytoplasmic polyadenylation element binding (CPEB) protein Orb2 as a positive regulator of rare-codon-dependent expression in neurons. Using RNA sequencing, we reveal that Orb2-upregulated mRNAs in the brain with abundant Orb2 binding sites have a rare-codon bias. From these Orb2-regulated mRNAs, we demonstrate that rare-codon enrichment is important for expression control and social behavior function of the metabotropic glutamate receptor (mGluR). Our findings reveal a molecular mechanism by which neural stem cell differentiation shifts genetic code regulation to enable critical mRNA and protein expression.
Collapse
|
12
|
Wei H, Du S, Parksong J, Pasolli HA, Matte-Martone C, Regot S, Gonzalez LE, Xin T, Greco V. Organ function is preserved despite reorganization of niche architecture in the hair follicle. Cell Stem Cell 2023; 30:962-972.e6. [PMID: 37419106 PMCID: PMC10362479 DOI: 10.1016/j.stem.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 05/01/2023] [Accepted: 06/07/2023] [Indexed: 07/09/2023]
Abstract
The ability of stem cells to build and replenish tissues depends on support from their niche. Although niche architecture varies across organs, its functional importance is unclear. During hair follicle growth, multipotent epithelial progenitors build hair via crosstalk with their remodeling fibroblast niche, the dermal papilla, providing a powerful model to functionally interrogate niche architecture. Through mouse intravital imaging, we show that dermal papilla fibroblasts remodel individually and collectively to form a morphologically polarized, structurally robust niche. Asymmetric TGF-β signaling precedes morphological niche polarity, and loss of TGF-β signaling in dermal papilla fibroblasts leads them to progressively lose their stereotypic architecture, instead surrounding the epithelium. The reorganized niche induces the redistribution of multipotent progenitors but nevertheless supports their proliferation and differentiation. However, the differentiated lineages and hairs produced by progenitors are shorter. Overall, our results reveal that niche architecture optimizes organ efficiency but is not absolutely essential for organ function.
Collapse
Affiliation(s)
- Haoyang Wei
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Shuangshuang Du
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jeeun Parksong
- Departments of Cell Biology and Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - H Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | | | - Sergi Regot
- Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Lauren E Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tianchi Xin
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Cell Biology and Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
13
|
Zion EH, Ringwalt D, Rinaldi K, Kahney EW, Li Y, Chen X. Old and newly synthesized histones are asymmetrically distributed in Drosophila intestinal stem cell divisions. EMBO Rep 2023; 24:e56404. [PMID: 37255015 PMCID: PMC10328082 DOI: 10.15252/embr.202256404] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/30/2023] [Accepted: 05/16/2023] [Indexed: 06/01/2023] Open
Abstract
We report that preexisting (old) and newly synthesized (new) histones H3 and H4 are asymmetrically partitioned during the division of Drosophila intestinal stem cells (ISCs). Furthermore, the inheritance patterns of old and new H3 and H4 in postmitotic cell pairs correlate with distinct expression patterns of Delta, an important cell fate gene. To understand the biological significance of this phenomenon, we expressed a mutant H3T3A to compromise asymmetric histone inheritance. Under this condition, we observe an increase in Delta-symmetric cell pairs and overpopulated ISC-like, Delta-positive cells. Single-cell RNA-seq assays further indicate that H3T3A expression compromises ISC differentiation. Together, our results indicate that asymmetric histone inheritance potentially contributes to establishing distinct cell identities in a somatic stem cell lineage, consistent with previous findings in Drosophila male germline stem cells.
Collapse
Affiliation(s)
- Emily H Zion
- Department of BiologyThe Johns Hopkins UniversityBaltimoreMDUSA
| | - Daniel Ringwalt
- Department of BiologyThe Johns Hopkins UniversityBaltimoreMDUSA
| | | | | | - Yingying Li
- Department of BiologyThe Johns Hopkins UniversityBaltimoreMDUSA
| | - Xin Chen
- Department of BiologyThe Johns Hopkins UniversityBaltimoreMDUSA
- Howard Hughes Medical InstituteBaltimoreMDUSA
| |
Collapse
|
14
|
Wodrich APK, Scott AW, Giniger E. What do we mean by "aging"? Questions and perspectives revealed by studies in Drosophila. Mech Ageing Dev 2023; 213:111839. [PMID: 37354919 PMCID: PMC10330756 DOI: 10.1016/j.mad.2023.111839] [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: 05/01/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
What is the nature of aging, and how best can we study it? Here, using a series of questions that highlight differing perspectives about the nature of aging, we ask how data from Drosophila melanogaster at the organismal, tissue, cellular, and molecular levels shed light on the complex interactions among the phenotypes associated with aging. Should aging be viewed as an individual's increasing probability of mortality over time or as a progression of physiological states? Are all age-correlated changes in physiology detrimental to vigor or are some compensatory changes that maintain vigor? Why do different age-correlated functions seem to change at different rates in a single individual as it ages? Should aging be considered as a single, integrated process across the scales of biological resolution, from organismal to molecular, or must we consider each level of biological scale as a separate, distinct entity? Viewing aging from these differing perspectives yields distinct but complementary interpretations about the properties and mechanisms of aging and may offer a path through the complexities related to understanding the nature of aging.
Collapse
Affiliation(s)
- Andrew P K Wodrich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, United States; Interdisciplinary Program in Neuroscience, Georgetown University, Washington DC, United States; College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Andrew W Scott
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, United States.
| |
Collapse
|
15
|
Galasso A, Xu DC, Hill C, Iakovleva D, Stefana MI, Baena‐Lopez LA. Non-apoptotic caspase activation ensures the homeostasis of ovarian somatic stem cells. EMBO Rep 2023; 24:e51716. [PMID: 37039000 PMCID: PMC10240206 DOI: 10.15252/embr.202051716] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/22/2023] [Accepted: 03/14/2023] [Indexed: 04/12/2023] Open
Abstract
Current evidence has associated caspase activation with the regulation of basic cellular functions without causing apoptosis. Malfunction of non-apoptotic caspase activities may contribute to specific neurological disorders, metabolic diseases, autoimmune conditions and cancers. However, our understanding of non-apoptotic caspase functions remains limited. Here, we show that non-apoptotic caspase activation prevents the intracellular accumulation of the Patched receptor in autophagosomes and the subsequent Patched-dependent induction of autophagy in Drosophila follicular stem cells. These events ultimately sustain Hedgehog signalling and the physiological properties of ovarian somatic stem cells and their progeny under moderate thermal stress. Importantly, our key findings are partially conserved in ovarian somatic cells of human origin. These observations attribute to caspases a pro-survival role under certain cellular conditions.
Collapse
Affiliation(s)
- Alessia Galasso
- Faculty of Medicine CentreImperial College London, South Kensington CampusLondonUK
| | - Derek Cui Xu
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Claire Hill
- School of Medicine, Dentistry and Biomedical SciencesQueen's University Belfast MedicineBelfastUK
| | - Daria Iakovleva
- Center for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | | | | |
Collapse
|
16
|
Moreno Acosta OD, Boan AF, Hattori RS, Fernandino JI. Notch pathway is required for protection against heat stress in spermatogonial stem cells in medaka. FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:487-500. [PMID: 37126120 DOI: 10.1007/s10695-023-01200-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 04/20/2023] [Indexed: 05/25/2023]
Abstract
Gamete production is a fundamental process for reproduction; however, exposure to stress, such as increased environmental temperature, can decrease or even interrupt this process, affecting fertility. Thus, the survival of spermatogonial stem cells (SSCs) is crucial for the recovery of spermatogenesis upon stressful situations. Here, we show that the Notch pathway is implicated in such survival, by protecting the SSCs against thermal stress. First, we corroborated the impairment of spermatogenesis under heat stress in medaka, observing an arrest in metaphase I at 10 days of heat treatment, an increase in the number of spermatocytes, and downregulation of ndrg1b and sycp3. In addition, at 30 days of treatment, an interruption of spermatogenesis was observed with a strong loss of spermatocytes and spermatids. Then, the exposure of adult males to thermal stress condition induced apoptosis mainly in spermatogenic and supporting somatic cells, with the exception of the germinal region, where SSCs are located. Concomitantly, the Notch pathway-related genes were upregulated, including the ligands (dll4, jag1-2) and receptors (notch1a-3). Moreover, during thermal stress presenilin enhancer-2 (pen-2), the catalytic subunit of γ-secretase complex of the Notch pathway was restricted to the germinal region of the medaka testis, observed in somatic cells surrounding type A spermatogonia (SGa). The importance of Notch pathway was further supported by an ex vivo approach, in which the inhibition of this pathway activity induced a loss of SSCs. Overall, this study supports the importance of Notch pathways for the protection of SSCs under chronic thermal stress.
Collapse
Affiliation(s)
- Omar D Moreno Acosta
- Instituto Tecnológico de Chascomús, INTECH (CONICET-UNSAM), Chascomus, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomus, Argentina
| | - Agustín F Boan
- Instituto Tecnológico de Chascomús, INTECH (CONICET-UNSAM), Chascomus, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomus, Argentina
| | - Ricardo S Hattori
- Salmonid Experimental Station at Campos Do Jordão, UPD-CJ, Sao Paulo Fisheries Institute (APTA/SAA), Campos Do Jordao, Brazil
| | - Juan Ignacio Fernandino
- Instituto Tecnológico de Chascomús, INTECH (CONICET-UNSAM), Chascomus, Argentina.
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomus, Argentina.
| |
Collapse
|
17
|
Abstract
In this chapter, we highlight examples of the diverse array of developmental, cellular, and biochemical insights that can be gained by using Drosophila melanogaster oogenesis as a model tissue. We begin with an overview of ovary development and adult oogenesis. Then we summarize how the adult Drosophila ovary continues to advance our understanding of stem cells, cell cycle, cell migration, cytoplasmic streaming, nurse cell dumping, and cell death. We also review emerging areas of study, including the roles of lipid droplets, ribosomes, and nuclear actin in egg development. Finally, we conclude by discussing the growing conservation of processes and signaling pathways that regulate oogenesis and female reproduction from flies to humans.
Collapse
|
18
|
Choi DY, Kim Y. PGE 2 mediation of egg development in Western flower thrip, Frankliniella occidentalis. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2023; 112:e21949. [PMID: 35749583 DOI: 10.1002/arch.21949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
Eicosanoids mediate various insect physiological processes, including reproduction. Especially, the eicosanoid prostaglandin E2 (PGE2 ) is known to mediate oocyte development in some insects. The explosive reproductive potential of the Western flower thrips, Frankliniella occidentalis, damages various agricultural crops. However, little is known about the underlying physiological processes of egg development in this pest. This study found that treatment with aspirin (ASP) (a specific cyclooxygenase (COX) inhibitor) used to inhibit PGE2 biosynthesis during ovarian development significantly suppressed the reproduction of female F. occidentalis. However, the addition of PGE2 to ASP-treated females significantly rescued the suppressed reproduction. PGE2 was detected in growing ovarian follicles in an immunofluorescence assay. The hypothetical biosynthetic machinery of PGE2 was predicted from the F. occidentalis genome and included phospholipase A2 (PLA2 ), COX-like peroxidase (POX), and PGE2 synthase (PGES). Three specific PLA2 s were highly expressed in female adults during active oogenesis. Specific POX and PGES genes also showed high expression during active oogenesis. The adverse effect of ASP treatment on oogenesis was observed in follicle formation in the germarium where the follicle numbers in an ovariole were decreased, which resulted in hypotrophied ovaries. This impairment was rescued by the addition of PGE2 . ASP treatment also significantly inhibited chorion formation and suppressed gene expression associated with choriogenesis, which included chorion protein, mucin, and yellow while it did not inhibit vitellogenin gene expression. However, the addition of PGE2 induced the expression of the target genes suppressed by ASP treatment and rescued chorion formation. These results suggest that PGE2 mediated ovarian development by affecting follicle formation and choriogenesis in F. occidentalis.
Collapse
Affiliation(s)
- Du-Yeol Choi
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, Korea
| | - Yonggyun Kim
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, Korea
| |
Collapse
|
19
|
Liu S, Baeg GH, Yang Y, Goh FG, Bao H, Wagner EJ, Yang X, Cai Y. The Integrator complex desensitizes cellular response to TGF-β/BMP signaling. Cell Rep 2023; 42:112007. [PMID: 36641752 DOI: 10.1016/j.celrep.2023.112007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/12/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Maintenance of stem cells requires the concerted actions of niche-derived signals and stem cell-intrinsic factors. Although Decapentaplegic (Dpp), a Drosophila bone morphogenetic protein (BMP) molecule, can act as a long-range morphogen, its function is spatially limited to the germline stem cell niche in the germarium. We show here that Integrator, a complex known to be involved in RNA polymerase II (RNAPII)-mediated transcriptional regulation in the nucleus, promotes germline differentiation by restricting niche-derived Dpp/BMP activity in the cytoplasm. Further results show that Integrator works in various developmental contexts to desensitize the cellular response to Dpp/BMP signaling during Drosophila development. Mechanistically, our results show that Integrator forms a multi-subunit complex with the type I receptor Thickveins (Tkv) and other Dpp/BMP signaling components and acts in a negative feedback loop to promote Tkv turnover independent of its transcriptional activity. Similarly, human Integrator subunits bind transforming growth factor β (TGF-β)/BMP signaling components and antagonize their activity, suggesting a conserved role of Integrator across metazoans.
Collapse
Affiliation(s)
- Sen Liu
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore
| | - Gyeong Hun Baeg
- Faculty of Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Ying Yang
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore
| | - Feng Guang Goh
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Hongcun Bao
- The Women's Hospital and Institute of Genetics, School of Medicine, Zhejiang University, Hang Zhou 310058, China
| | - Eric J Wagner
- Department of Biochemistry and Biophysics, Center for RNA Biology, Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, KMRB B.9629, Rochester, NY 14642 USA
| | - Xiaohang Yang
- The Women's Hospital and Institute of Genetics, School of Medicine, Zhejiang University, Hang Zhou 310058, China
| | - Yu Cai
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.
| |
Collapse
|
20
|
Zhang J, Zhang S, Sun Z, Cai Y, Zhong G, Yi X. Camptothecin Effectively Regulates Germline Differentiation through Bam-Cyclin A Axis in Drosophila melanogaster. Int J Mol Sci 2023; 24:ijms24021617. [PMID: 36675143 PMCID: PMC9864452 DOI: 10.3390/ijms24021617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Camptothecin (CPT), first isolated from Chinese tree Camptotheca acuminate, produces rapid and prolonged inhibition of DNA synthesis and induction of DNA damage by targeting topoisomerase I (top1), which is highly activated in cancer cells. CPT thus exhibits remarkable anticancer activities in various cancer types, and is a promising therapeutic agent for the treatment of cancers. However, it remains to be uncovered underlying its cytotoxicity toward germ cells. In this study we found that CPT, a cell cycle-specific anticancer agent, reduced fecundity and exhibited significant cytotoxicity toward GSCs and two-cell cysts. We showed that CPT induced GSC loss and retarded two-cell cysts differentiation in a niche- or apoptosis-independent manner. Instead, CPT induced ectopic expression of a differentiation factor, bag of marbles (Bam), and regulated the expression of cyclin A, which contributed to GSC loss. In addition, CPT compromised two-cell cysts differentiation by decreasing the expression of Bam and inducing cell arrest at G1/S phase via cyclin A, eventually resulting in two-cell accumulation. Collectively, this study demonstrates, for the first time in vivo, that the Bam-cyclin A axis is involved in CPT-mediated germline stem cell loss and two-cell cysts differentiation defects via inducing cell cycle arrest, which could provide information underlying toxicological effects of CPT in the productive system, and feature its potential to develop as a pharmacology-based germline stem cell regulation agent.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Shijie Zhang
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Zhipeng Sun
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Yu Cai
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 119077, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore 119077, Singapore
| | - Guohua Zhong
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (G.Z.); (X.Y.)
| | - Xin Yi
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (G.Z.); (X.Y.)
| |
Collapse
|
21
|
Cai Q, Yan J, Duan R, Zhu Y, Hua Y, Liao Y, Li Q, Li W, Ji S. E3 ligase Cul2 mediates Drosophila early germ cell differentiation through targeting Bam. Dev Biol 2023; 493:103-108. [PMID: 36423673 DOI: 10.1016/j.ydbio.2022.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Drosophila ovary has been one of the most mature and excellent systems for studying the in vivo regulatory mechanisms of stem cell fate determination. It has been well-known that the bone morphogenetic protein (BMP) signaling released by the niche cells promotes the maintenance of germline stem cells (GSCs) through inhibiting the transcription of the bag-of-marbles (bam) gene, which encodes a key factor for GSC differentiation. However, whether Bam is regulated at the post-translational level remains largely unknown. Here we show that the E3 ligase Cullin-2 (Cul2) is involved in modulating Bam ubiquitination, which occurs probably at multiple lysine residues of Bam's C-terminal region. Genetic evidence further supports the notion that Cul2-mediated Bam ubiquitination and turnover are essential for GSC maintenance and proper germline development. Collectively, our data not only uncovers a novel regulatory mechanism by which Bam is controlled at the post-translational level, but also provides new insights into how Cullin family protein determines the differentiation fate of early germ cells.
Collapse
Affiliation(s)
- Qingshuang Cai
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Jing Yan
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Renjie Duan
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Yangyang Zhu
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Yongzhi Hua
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Yongrong Liao
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Qingyang Li
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Weini Li
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Shanming Ji
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China.
| |
Collapse
|
22
|
Chatterjee D, Deng WM. Standardization of Single-Cell RNA-Sequencing Analysis Workflow to Study Drosophila Ovary. Methods Mol Biol 2023; 2677:151-171. [PMID: 37464241 DOI: 10.1007/978-1-0716-3259-8_9] [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: 07/20/2023]
Abstract
Developments in single-cell technology have considerably changed the way we study biology. Significant efforts have been made over the last few years to build comprehensive cell-type-specific transcriptomic atlases for a wide range of tissues in several model organisms in order to discover cell-type-specific markers and drivers of gene expression. One such tissue is the ovary of the fruit-fly Drosophila melanogaster, which is a popular model system with wide-ranging applications in the study of both development and disease. Three independent studies have recently produced comprehensive maps of cell-type-specific gene expression that describe both spatiotemporal regulation of the process of oogenesis and unique transcriptomic profiles of different cell types that constitute the ovary. In this chapter, we outlined the wet-lab protocol that was followed in our recent study for sample preparation and reanalyze the resultant dataset to discuss the benchmarks in data analysis, which are fundamental to comprehensive curation of the single-cell dataset representing the fly ovary.
Collapse
Affiliation(s)
- Deeptiman Chatterjee
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA.
- Current address: Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA, USA.
| |
Collapse
|
23
|
Yao Y, Gu X, Xu X, Ge S, Jia R. Novel insights into RB1 mutation. Cancer Lett 2022; 547:215870. [PMID: 35964818 DOI: 10.1016/j.canlet.2022.215870] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 01/09/2023]
Abstract
Since the discovery of the retinoblastoma susceptibility gene (RB1) decades ago, RB1 has been regarded as a prototype tumor suppressor gene providing a paradigm for tumor genetic research. Constant research has updated the understanding of RB1-related pathways and their impact on tumor and nontumor diseases. Mutation of RB1 gene has been observed in multiple types of malignant tumors including prostate cancer, lung cancer, breast cancer, and almost every familial and sporadic case of retinoblastoma. Even if well-known and long-investigated, the application potential of RB1 mutation has not been fully tapped. In this review, we focus on the mechanism underlying RB1 mutation during oncogenesis. Therapeutically, we have further discussed potential clinical strategies by targeting RB1-mutated cancers. The unsolved problems and prospects of RB1 mutation are also discussed.
Collapse
Affiliation(s)
- Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Xiaofang Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
24
|
Zhao H, Li Z, Kong R, Shi L, Ma R, Ren X, Li Z. Novel intrinsic factor Yun maintains female germline stem cell fate through Thickveins. Stem Cell Reports 2022; 17:1914-1923. [PMID: 35985332 PMCID: PMC9481913 DOI: 10.1016/j.stemcr.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/10/2022] Open
Abstract
Germline stem cells (GSCs) are critical for the reproduction of an organism. The self-renewal and differentiation of GSCs must be tightly controlled to avoid uncontrolled stem cell proliferation or premature stem cell differentiation. However, how the self-renewal and differentiation of GSCs are properly controlled is not fully understood. Here, we find that the novel intrinsic factor Yun is required for female GSC maintenance in Drosophila. GSCs undergo precocious differentiation due to de-repression of differentiation factor Bam by defective BMP/Dpp signaling in the absence of yun. Mechanistically, Yun associates with and stabilizes Thickveins (Tkv), the type I receptor of Dpp/BMP signaling. Finally, ectopic expression of a constitutively active Tkv (TkvQD) completely suppresses GSC loss caused by yun depletion. Collectively, these data demonstrate that Yun functions through Tkv to maintain GSC fate. Our results provide new insight into the regulatory mechanisms of how stem cell maintenance is properly controlled. Novel intrinsic factor Yun is required for female GSC maintenance Yun-defective GSCs undergo differentiation due to Bam upregulation Yun associates with and stabilizes Tkv to regulate GSC maintenance GSC loss in the absence of yun could be rescued by constitutively active Tkv
Collapse
Affiliation(s)
- Hang Zhao
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Zhengran Li
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Ruiyan Kong
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Lin Shi
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Rui Ma
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xuejing Ren
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing 100048, China.
| |
Collapse
|
25
|
Diegmiller R, Nunley H, Shvartsman SY, Imran Alsous J. Quantitative models for building and growing fated small cell networks. Interface Focus 2022; 12:20210082. [PMID: 35865502 PMCID: PMC9184967 DOI: 10.1098/rsfs.2021.0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
Small cell clusters exhibit numerous phenomena typically associated with complex systems, such as division of labour and programmed cell death. A conserved class of such clusters occurs during oogenesis in the form of germline cysts that give rise to oocytes. Germline cysts form through cell divisions with incomplete cytokinesis, leaving cells intimately connected through intercellular bridges that facilitate cyst generation, cell fate determination and collective growth dynamics. Using the well-characterized Drosophila melanogaster female germline cyst as a foundation, we present mathematical models rooted in the dynamics of cell cycle proteins and their interactions to explain the generation of germline cell lineage trees (CLTs) and highlight the diversity of observed CLT sizes and topologies across species. We analyse competing models of symmetry breaking in CLTs to rationalize the observed dynamics and robustness of oocyte fate specification, and highlight remaining gaps in knowledge. We also explore how CLT topology affects cell cycle dynamics and synchronization and highlight mechanisms of intercellular coupling that underlie the observed collective growth patterns during oogenesis. Throughout, we point to similarities across organisms that warrant further investigation and comment on the extent to which experimental and theoretical findings made in model systems extend to other species.
Collapse
Affiliation(s)
- Rocky Diegmiller
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Hayden Nunley
- Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Stanislav Y. Shvartsman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA,Department of Molecular Biology, Princeton University, Princeton, NJ, USA,Flatiron Institute, Simons Foundation, New York, NY, USA
| | | |
Collapse
|
26
|
Ariyapala IS, Buddika K, Hundley HA, Calvi BR, Sokol NS. The RNA binding protein Swm is critical for Drosophila melanogaster intestinal progenitor cell maintenance. Genetics 2022; 222:6619166. [PMID: 35762963 DOI: 10.1093/genetics/iyac099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
The regulation of stem cell survival, self-renewal, and differentiation is critical for the maintenance of tissue homeostasis. Although the involvement of signaling pathways and transcriptional control mechanisms in stem cell regulation have been extensively investigated, the role of post-transcriptional control is still poorly understood. Here we show that the nuclear activity of the RNA-binding protein Second Mitotic Wave Missing (Swm) is critical for Drosophila melanogaster intestinal stem cells (ISCs) and their daughter cells, enteroblasts (EBs), to maintain their progenitor cell properties and functions. Loss of swm causes ISCs and EBs to stop dividing and instead detach from the basement membrane, resulting in severe progenitor cell loss. swm loss is further characterized by nuclear accumulation of poly(A)+ RNA in progenitor cells. Swm associates with transcripts involved in epithelial cell maintenance and adhesion, and the loss of swm, while not generally affecting the levels of these Swm-bound mRNAs, leads to elevated expression of proteins encoded by some of them, including the fly ortholog of Filamin. Taken together, this study indicates a nuclear role for Swm in adult stem cell maintenance, raising the possibility that nuclear post-transcriptional regulation of mRNAs encoding cell adhesion proteins ensures proper attachment of progenitor cells.
Collapse
Affiliation(s)
| | - Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Heather A Hundley
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Brian R Calvi
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
27
|
Anllo L, DiNardo S. Visceral mesoderm signaling regulates assembly position and function of the Drosophila testis niche. Dev Cell 2022; 57:1009-1023.e5. [PMID: 35390292 PMCID: PMC9050945 DOI: 10.1016/j.devcel.2022.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022]
Abstract
Tissue homeostasis often requires a properly placed niche to support stem cells. Morphogenetic processes that position a niche are just being described. For the Drosophila testis, we recently showed that pro-niche cells, specified at disparate positions during early gonadogenesis, must assemble into one collective at the anterior of the gonad. We now find that Slit and FGF signals emanating from adjacent visceral mesoderm regulate assembly. In response to signaling, niche cells express islet, which we find is also required for niche assembly. Without signaling, niche cells specified furthest from the anterior are unable to migrate, remaining dispersed. The function of such niches is severely disrupted, with niche cells evading cell cycle quiescence, compromised in their ability to signal the incipient stem cell pool, and failing to orient stem cell divisions properly. Our work identifies both extrinsic signaling and intrinsic responses required for proper assembly and placement of the testis niche.
Collapse
Affiliation(s)
- Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| |
Collapse
|
28
|
Patel A, Wu Y, Han X, Su Y, Maugel T, Shroff H, Roy S. Cytonemes coordinate asymmetric signaling and organization in the Drosophila muscle progenitor niche. Nat Commun 2022; 13:1185. [PMID: 35246530 PMCID: PMC8897416 DOI: 10.1038/s41467-022-28587-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/02/2022] [Indexed: 12/29/2022] Open
Abstract
Asymmetric signaling and organization in the stem-cell niche determine stem-cell fates. Here, we investigate the basis of asymmetric signaling and stem-cell organization using the Drosophila wing-disc that creates an adult muscle progenitor (AMP) niche. We show that AMPs extend polarized cytonemes to contact the disc epithelial junctions and adhere themselves to the disc/niche. Niche-adhering cytonemes localize FGF-receptor to selectively adhere to the FGF-producing disc and receive FGFs in a contact-dependent manner. Activation of FGF signaling in AMPs, in turn, reinforces disc-specific cytoneme polarity/adhesion, which maintains their disc-proximal positions. Loss of cytoneme-mediated adhesion promotes AMPs to lose niche occupancy and FGF signaling, occupy a disc-distal position, and acquire morphological hallmarks of differentiation. Niche-specific AMP organization and diversification patterns are determined by localized expression and presentation patterns of two different FGFs in the wing-disc and their polarized target-specific distribution through niche-adhering cytonemes. Thus, cytonemes are essential for asymmetric signaling and niche-specific AMP organization.
Collapse
Affiliation(s)
- Akshay Patel
- grid.164295.d0000 0001 0941 7177Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD USA
| | - Yicong Wu
- grid.94365.3d0000 0001 2297 5165Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA
| | - Xiaofei Han
- grid.94365.3d0000 0001 2297 5165Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA
| | - Yijun Su
- grid.94365.3d0000 0001 2297 5165Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA ,grid.94365.3d0000 0001 2297 5165Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD USA
| | - Tim Maugel
- grid.164295.d0000 0001 0941 7177Department of Biology, Laboratory for Biological Ultrastructure, University of Maryland, College Park, MD USA
| | - Hari Shroff
- grid.94365.3d0000 0001 2297 5165Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA ,grid.94365.3d0000 0001 2297 5165Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD USA
| | - Sougata Roy
- grid.164295.d0000 0001 0941 7177Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD USA
| |
Collapse
|
29
|
To V, Kim HJ, Jang W, Sreejith P, Kim C. Lin28 and Imp are Required for Stability of Bowl Transcripts in Hub
Cells of the Drosophila Testis. Dev Reprod 2021; 25:313-319. [PMID: 35141457 PMCID: PMC8807131 DOI: 10.12717/dr.2021.25.4.313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/28/2021] [Accepted: 12/15/2021] [Indexed: 11/24/2022]
Abstract
Hub cells comprise a niche for germline stem cells and cyst stem cells in the
Drosophila testis. Hub cells arise from common somatic
gonadal precursors in embryos, but the mechanism of their specification is still
poorly understood. Here we find that RNA binding proteins Lin28 and Imp mediate
transcript stability of Bowl, a known hub specification factor; Bowl transcripts
were reduced in the testis of Lin28 and Imp mutants, and also when RNA-mediated
interference against Lin28 or Imp was expressed in hub cells. In tissue culture
Luciferase assays involving the Bowl 3’UTR, stability of Luc reporter
transcripts depended on the Bowl 3’UTR and required Lin28 and Imp. Our
findings suggest that proper Bowl function during hub cell specification
requires Lin28 and Imp in the testis hub cells.
Collapse
Affiliation(s)
- Van To
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | - Hyun Ju Kim
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | - Wijeong Jang
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | | | - Changsoo Kim
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
- Corresponding author Changsoo Kim, School of
Biological Sciences and Technology, Chonnam National University, Gwangju 61186,
Korea. Tel: +82-62-530-5201, E-mail:
| |
Collapse
|
30
|
Jangid A, Selvarajan S, Ramaswamy R. A stochastic model of homeostasis: The roles of noise and nuclear positioning in deciding cell fate. iScience 2021; 24:103199. [PMID: 34703995 PMCID: PMC8524154 DOI: 10.1016/j.isci.2021.103199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/21/2021] [Accepted: 09/28/2021] [Indexed: 11/27/2022] Open
Abstract
We study a population-based cellular model that starts from a single stem cell that divides stochastically to give rise to either daughter stem cells or differentiated daughter cells. There are three main components in the model: nucleus position, the underlying gene-regulatory network, and stochastic segregation of transcription factors in the daughter cells. The proportion of self-renewal and differentiated cell lines as a function of the nucleus position which in turn decides the plane of cleavage is studied. Both nuclear position and noise play an important role in determining the stem cell genealogies. We have observed both long and short genealogies in model simulation, and these compare well with experimental results from neuroblast and B-cell division. Symmetric divisions are observed in apical nuclei, while asymmetric division occurs when the nucleus is toward the base. In this model, the number of clones decreases over time, although the average clone size increases.
Collapse
Affiliation(s)
- Amit Jangid
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Suriya Selvarajan
- Department of Theoretical Physics, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Ram Ramaswamy
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| |
Collapse
|
31
|
Reilein A, Kogan HV, Misner R, Park KS, Kalderon D. Adult stem cells and niche cells segregate gradually from common precursors that build the adult Drosophila ovary during pupal development. eLife 2021; 10:69749. [PMID: 34590579 PMCID: PMC8536258 DOI: 10.7554/elife.69749] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 09/29/2021] [Indexed: 12/31/2022] Open
Abstract
Production of proliferative follicle cells (FCs) and quiescent escort cells (ECs) by follicle stem cells (FSCs) in adult Drosophila ovaries is regulated by niche signals from anterior (cap cells, ECs) and posterior (polar FCs) sources. Here we show that ECs, FSCs, and FCs develop from common pupal precursors, with different fates acquired by progressive separation of cells along the AP axis and a graded decline in anterior cell proliferation. ECs, FSCs, and most FCs derive from intermingled cell (IC) precursors interspersed with germline cells. Precursors also accumulate posterior to ICs before engulfing a naked germline cyst projected out of the germarium to form the first egg chamber and posterior polar FC signaling center. Thus, stem and niche cells develop in appropriate numbers and spatial organization through regulated proliferative expansion together with progressive establishment of spatial signaling cues that guide adult cell behavior, rather than through rigid early specification events.
Collapse
Affiliation(s)
- Amy Reilein
- Department of Biological Sciences, Columbia University, New York, United States
| | - Helen V Kogan
- Department of Biological Sciences, Columbia University, New York, United States
| | - Rachel Misner
- Department of Biological Sciences, Columbia University, New York, United States
| | - Karen Sophia Park
- Department of Biological Sciences, Columbia University, New York, United States
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, United States
| |
Collapse
|
32
|
Goutas A, Trachana V. Stem cells' centrosomes: How can organelles identified 130 years ago contribute to the future of regenerative medicine? World J Stem Cells 2021; 13:1177-1196. [PMID: 34630857 PMCID: PMC8474719 DOI: 10.4252/wjsc.v13.i9.1177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/03/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023] Open
Abstract
At the core of regenerative medicine lies the expectation of repair or replacement of damaged tissues or whole organs. Donor scarcity and transplant rejection are major obstacles, and exactly the obstacles that stem cell-based therapy promises to overcome. These therapies demand a comprehensive understanding of the asymmetric division of stem cells, i.e. their ability to produce cells with identical potency or differentiated cells. It is believed that with better understanding, researchers will be able to direct stem cell differentiation. Here, we describe extraordinary advances in manipulating stem cell fate that show that we need to focus on the centrosome and the centrosome-derived primary cilium. This belief comes from the fact that this organelle is the vehicle that coordinates the asymmetric division of stem cells. This is supported by studies that report the significant role of the centrosome/cilium in orchestrating signaling pathways that dictate stem cell fate. We anticipate that there is sufficient evidence to place this organelle at the center of efforts that will shape the future of regenerative medicine.
Collapse
Affiliation(s)
- Andreas Goutas
- Department of Biology, Faculty of Medicine, University of Thessaly, Larisa 41500, Biopolis, Greece
| | - Varvara Trachana
- Department of Biology, Faculty of Medicine, University of Thessaly, Larisa 41500, Biopolis, Greece.
| |
Collapse
|
33
|
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer that can self-renew and differentiate into large tumor masses. Evidence accumulated to date shows that CSCs affect tumor proliferation, recurrence, and resistance to chemotherapy. Recent studies have shown that, like stem cells, CSCs maintain cells with self-renewal capacity by means of asymmetric division and promote cell proliferation by means of symmetric division. This cell division is regulated by fate determinants, such as the NUMB protein, which recently has also been confirmed as a tumor suppressor. Loss of NUMB expression leads to uncontrolled proliferation and amplification of the CSC pool, which promotes the Notch signaling pathway and reduces the expression of the p53 protein. NUMB genes are alternatively spliced to produce six functionally distinct isoforms. An interesting recent discovery is that the protein NUMB isoform produced by alternative splicing of NUMB plays an important role in promoting carcinogenesis. In this review, we summarize the known functions of NUMB and NUMB isoforms related to the proliferation and generation of CSCs.
Collapse
Affiliation(s)
- Hye Yeon Choi
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA
| | - Jaekwon Seok
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University, Seoul 05029, Korea
| | - Geun-Ho Kang
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University, Seoul 05029, Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University, Seoul 05029, Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University, Seoul 05029, Korea
| |
Collapse
|
34
|
Xu C, Ding YH, Wang K, Hao M, Li H, Ding L. Claudin-7 deficiency promotes stemness properties in colorectal cancer through Sox9-mediated Wnt/β-catenin signalling. J Transl Med 2021; 19:311. [PMID: 34281572 PMCID: PMC8287764 DOI: 10.1186/s12967-021-02983-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/11/2021] [Indexed: 02/08/2023] Open
Abstract
Background Colorectal cancer (CRC) is a common malignant tumour of the digestive tract that is characterized by high patient morbidity and mortality rates. Claudin-7 (Cldn7), a tight junction protein, was recently reported to function as a candidate tumour suppressor gene in CRC. Our previous study demonstrated that the large intestine of C57/BL6 mice showed intestinal adenomas and abnormal Ki67 expression and distribution in the intestinal crypt when Cldn7 was knocked out. The aim of this study was to further investigate whether Cldn7 deficiency has non-tight junction functions, affects intestinal stemness properties, promotes CRC and to determine the specific mechanism. Methods Cell proliferation assays, migration assays, apoptosis assays, tumour sphere formation assays in vitro, and subcutaneous xenograft models in vivo were used to determine the effects of Cldn7 knockdown on the biological characteristics of CRC stem cells. Western blotting, qPCR and immunofluorescence staining were performed to identify the epithelial-mesenchymal transition and the activation of Wnt/β-catenin pathway in CRC stem cells. Cldn7 inducible conditional gene knockout mice and immunohistochemical staining further verified this hypothesis in vivo. The mechanism and target of Cldn7 were determined by performing a chromatin immunoprecipitation (ChIP) assay and coimmunoprecipitation (CoIP) assay. Results Cldn7 knock down in CRC stem cells promoted cell proliferation, migration, and globular growth in serum-free medium and the ability to form xenograft tumours; cell apoptosis was inhibited, while the cellular epithelial-mesenchymal transition was also observed. These changes in cell characteristics were achieved by activating the Wnt/β-catenin pathway and promoting the expression of downstream target genes after β-catenin entry into the nucleus, as observed in CRC cell lines and Cldn7 gene knockout mouse experiments. Using ChIP and CoIP experiments, we initially found that Cldn7 and Sox9 interacted at the protein level to activate the Wnt/β-catenin pathway. Conclusions Based on our research, Cldn7 deficiency confers stemness properties in CRC through Sox9-mediated Wnt/β-catenin signalling. This result clarifies that Cldn7 plays an inhibitory role in CRC and reveals a possible molecular mechanism, which is conducive to further research on Cldn7 and cancer stem cells.
Collapse
Affiliation(s)
- Chang Xu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.,Department of Hepato-Pancreato-Biliary Surgery, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing , Peking University Cancer Hospital and Institute, Beijing, China
| | - Yu-Han Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Kun Wang
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Mengdi Hao
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Huimin Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Lei Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
| |
Collapse
|
35
|
Choi HY, Seok J, Kang GH, Lim KM, Cho SG. The role of NUMB/NUMB isoforms in cancer stem cells. BMB Rep 2021; 54:335-343. [PMID: 34078527 PMCID: PMC8328821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2023] Open
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer that can self-renew and differentiate into large tumor masses. Evidence accumulated to date shows that CSCs affect tumor proliferation, recurrence, and resistance to chemotherapy. Recent studies have shown that, like stem cells, CSCs maintain cells with self-renewal capacity by means of asymmetric division and promote cell proliferation by means of symmetric division. This cell division is regulated by fate determinants, such as the NUMB protein, which recently has also been confirmed as a tumor suppressor. Loss of NUMB expression leads to uncontrolled proliferation and amplification of the CSC pool, which promotes the Notch signaling pathway and reduces the expression of the p53 protein. NUMB genes are alternatively spliced to produce six functionally distinct isoforms. An interesting recent discovery is that the protein NUMB isoform produced by alternative splicing of NUMB plays an important role in promoting carcinogenesis. In this review, we summarize the known functions of NUMB and NUMB isoforms related to the proliferation and generation of CSCs. [BMB Reports 2021; 54(7): 335-343].
Collapse
Affiliation(s)
- Hye Yeon Choi
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA 90033, USA, Seoul 05029, Korea
| | - Jaekwon Seok
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University, Seoul 05029, Korea
| | - Geun-Ho Kang
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University, Seoul 05029, Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University, Seoul 05029, Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center (MCRC), Konkuk University, Seoul 05029, Korea
| |
Collapse
|
36
|
Hainaut M, Clarke HJ. Germ cells of the mammalian female: A limited or renewable resource? Biol Reprod 2021; 105:774-788. [PMID: 34114006 DOI: 10.1093/biolre/ioab115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 11/12/2022] Open
Abstract
In many non-mammalian organisms, a population of germ-line stem cells supports continuing production of gametes during most or all the life of the individual, and germ-line stem cells are also present and functional in male mammals. Traditionally, however, they have been thought not to exist in female mammals, who instead generate all their germ cells during fetal life. Over the last several years, this dogma has been challenged by several reports, while supported by others. We describe and compare these conflicting studies with the aim of understanding how they came to opposing conclusions. We first consider studies that, by examining marker-gene expression, the fate of genetically marked cells, and consequences of depleting the oocyte population, addressed whether ovaries of post-natal females contain oogonial stem cells (OSC) that give rise to new oocytes. We next discuss whether ovaries contain cells that, even if inactive under physiological conditions, nonetheless possess OSC properties that can be revealed through cell-culture. We then examine studies of whether cells harvested after long-term culture of cells obtained from ovaries can, following transplantation into ovaries of recipient females, give rise to oocytes and offspring. Finally, we note studies where somatic cells have been re-programmed to acquire a female germ-cell fate. We conclude that the weight of evidence strongly supports the traditional interpretation that germ-line stem cells do not exist post-natally in female mammals. However, the ability to generate germ cells from somatic cells in vitro establishes a method to generate new gametes from cells of post-natal mammalian females.
Collapse
Affiliation(s)
- Mathilde Hainaut
- Department of Obstetrics and Gynecology, McGill University and Research Institute of the McGill University Health Centre, Montreal Canada
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University and Research Institute of the McGill University Health Centre, Montreal Canada
| |
Collapse
|
37
|
Tiwari SK, Mandal S. Mitochondrial Control of Stem Cell State and Fate: Lessons From Drosophila. Front Cell Dev Biol 2021; 9:606639. [PMID: 34012959 PMCID: PMC8128071 DOI: 10.3389/fcell.2021.606639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/06/2021] [Indexed: 01/09/2023] Open
Abstract
Over the years, Drosophila has served as a wonderful genetically tractable model system to unravel various facets of tissue-resident stem cells in their microenvironment. Studies in different stem and progenitor cell types of Drosophila have led to the discovery of cell-intrinsic and extrinsic factors crucial for stem cell state and fate. Though initially touted as the ATP generating machines for carrying various cellular processes, it is now increasingly becoming clear that mitochondrial processes alone can override the cellular program of stem cells. The last few years have witnessed a surge in our understanding of mitochondria's contribution to governing different stem cell properties in their subtissular niches in Drosophila. Through this review, we intend to sum up and highlight the outcome of these in vivo studies that implicate mitochondria as a central regulator of stem cell fate decisions; to find the commonalities and uniqueness associated with these regulatory mechanisms.
Collapse
Affiliation(s)
- Satish Kumar Tiwari
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| | - Sudip Mandal
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| |
Collapse
|
38
|
Gong Y, Alassimone J, Varnau R, Sharma N, Cheung LS, Bergmann DC. Tuning self-renewal in the Arabidopsis stomatal lineage by hormone and nutrient regulation of asymmetric cell division. eLife 2021; 10:e63335. [PMID: 33739283 PMCID: PMC8009662 DOI: 10.7554/elife.63335] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/18/2021] [Indexed: 02/03/2023] Open
Abstract
Asymmetric and self-renewing divisions build and pattern tissues. In the Arabidopsis stomatal lineage, asymmetric cell divisions, guided by polarly localized cortical proteins, generate most cells on the leaf surface. Systemic and environmental signals modify tissue development, but the mechanisms by which plants incorporate such cues to regulate asymmetric divisions are elusive. In a screen for modulators of cell polarity, we identified CONSTITUTIVE TRIPLE RESPONSE1, a negative regulator of ethylene signaling. We subsequently revealed antagonistic impacts of ethylene and glucose signaling on the self-renewing capacity of stomatal lineage stem cells. Quantitative analysis of cell polarity and fate dynamics showed that developmental information may be encoded in both the spatial and temporal asymmetries of polarity proteins. These results provide a framework for a mechanistic understanding of how nutritional status and environmental factors tune stem-cell behavior in the stomatal lineage, ultimately enabling flexibility in leaf size and cell-type composition.
Collapse
Affiliation(s)
- Yan Gong
- Department of Biology, Stanford UniversityStanfordUnited States
| | | | - Rachel Varnau
- Department of Biology, Stanford UniversityStanfordUnited States
| | - Nidhi Sharma
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Lily S Cheung
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Dominique C Bergmann
- Department of Biology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
39
|
Wnt6 regulates the homeostasis of the stem cell niche via Rac1-and Cdc42-mediated noncanonical Wnt signalling pathways in Drosophila testis. Exp Cell Res 2021; 402:112511. [PMID: 33582096 DOI: 10.1016/j.yexcr.2021.112511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/18/2021] [Accepted: 01/26/2021] [Indexed: 11/23/2022]
Abstract
The homeostasis of the stem cell niche is regulated by both intrinsic and extrinsic factors, and the complex and ordered molecular and cellular regulatory mechanisms need to be further explored. In Drosophila testis, germline stem cells (GSCs) rely on hub cells for self-renewal and physical attachment. GSCs are also in contact with somatic cyst stem cells (CySCs). Utilizing genetic manipulation in Drosophila, we investigated the role of Wnt6 in vivo and in vitro. In Drosophila testis, we found that Wnt6 is required for GSC differentiation and CySC self-renewal. In Schneider 2 (S2) cells, we found that Wnt6 regulates cell proliferation and apoptosis. Mechanistically, we demonstrated that Wnt6 can downregulate the expression levels of Arm, Rac1 and Cdc42 in S2 cells. Notably, Rac1 and Cdc42, which act downstream of the noncanonical Wnt signalling pathway, imitated the phenotypes of Wnt6 in Drosophila testis. Thus, the newly discovered Wnt6-Rac1/Cdc42 signal axis is required for the homeostasis of the stem cell niche in the Drosophila testis.
Collapse
|
40
|
Centromere assembly and non-random sister chromatid segregation in stem cells. Essays Biochem 2021; 64:223-232. [PMID: 32406510 DOI: 10.1042/ebc20190066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/21/2020] [Accepted: 04/30/2020] [Indexed: 01/17/2023]
Abstract
Asymmetric cell division (ACD) produces daughter cells with separate distinct cell fates and is critical for the development and regulation of multicellular organisms. Epigenetic mechanisms are key players in cell fate determination. Centromeres, epigenetically specified loci defined by the presence of the histone H3-variant, centromere protein A (CENP-A), are essential for chromosome segregation at cell division. ACDs in stem cells and in oocyte meiosis have been proposed to be reliant on centromere integrity for the regulation of the non-random segregation of chromosomes. It has recently been shown that CENP-A is asymmetrically distributed between the centromeres of sister chromatids in male and female Drosophila germline stem cells (GSCs), with more CENP-A on sister chromatids to be segregated to the GSC. This imbalance in centromere strength correlates with the temporal and asymmetric assembly of the mitotic spindle and potentially orientates the cell to allow for biased sister chromatid retention in stem cells. In this essay, we discuss the recent evidence for asymmetric sister centromeres in stem cells. Thereafter, we discuss mechanistic avenues to establish this sister centromere asymmetry and how it ultimately might influence cell fate.
Collapse
|
41
|
Nandi A, Chowdhuri DK. Cadmium mediated redox modulation in germline stem cells homeostasis affects reproductive health of Drosophila males. JOURNAL OF HAZARDOUS MATERIALS 2021; 402:123737. [PMID: 33254766 DOI: 10.1016/j.jhazmat.2020.123737] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 06/12/2023]
Abstract
Maintenance of male germline stem cells (GSCs) homeostasis is crucial for successful reproductive life of adults. New insights gained on dysfunction in stem cell maintenance could be the basis of stem cell dependent ailment during adulthood. Cadmium (Cd), a reported male reproductive toxicant, has been explored inadequately for its impact on male GSCs maintenance. The present study, therefore, has been aimed to evaluate the adverse effect of Cd on the homeostasis of GSCs by using Drosophila testis as an in vivo model. Following developmental exposure of environmentally relevant concentrations of Cd (5.0, 10.0 and 20.0 μg/mL) to Drosophila, we showed that a significantly increased level of reactive oxygen species (ROS) at 20.0 μg/mL of Cd resulted in alteration of GSCs number accompanied by inappropriate differentiation leading to reduced sperm number and eventually poor reproductive performance in exposed organism. Rescuing effect was evident by overexpressing sod in the early germ cell stage. The study suggests that an alteration in GSCs homeostasis due to redox imbalance plays a pivotal role in Cd induced failure in male fertility. The study further advocates for the use of Drosophila as an alternative animal model for in vivo evaluation of male GSCs toxicity with minimal ethical concern.
Collapse
Affiliation(s)
- Abik Nandi
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, Uttar Pradesh, India
| | - Debapratim Kar Chowdhuri
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, Uttar Pradesh, India.
| |
Collapse
|
42
|
Abstract
The centrosome is a unique organelle: the semi-conservative nature of its duplication generates an inherent asymmetry between ‘mother’ and ‘daughter’ centrosomes, which differ in their age. This asymmetry has captivated many cell biologists, but its meaning has remained enigmatic. In the last two decades, many stem cell types have been shown to display stereotypical inheritance of either the mother or daughter centrosome. These observations have led to speculation that the mother and daughter centrosomes bear distinct information, contributing to differential cell fates during asymmetric cell divisions. This review summarizes recent progress and discusses how centrosome asymmetry may promote asymmetric fates during stem cell divisions.
Collapse
Affiliation(s)
- Cuie Chen
- Life Sciences Institute, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yukiko M Yamashita
- Life Sciences Institute, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.,Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, Howard Hughes Medical Institute, Cambridge, MA, USA
| |
Collapse
|
43
|
Tu R, Duan B, Song X, Chen S, Scott A, Hall K, Blanck J, DeGraffenreid D, Li H, Perera A, Haug J, Xie T. Multiple Niche Compartments Orchestrate Stepwise Germline Stem Cell Progeny Differentiation. Curr Biol 2020; 31:827-839.e3. [PMID: 33357404 DOI: 10.1016/j.cub.2020.12.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 11/17/2020] [Accepted: 12/15/2020] [Indexed: 11/28/2022]
Abstract
The niche controls stem cell self-renewal and progenitor differentiation for maintaining adult tissue homeostasis in various organisms. However, it remains unclear whether the niche is compartmentalized to control stem cell self-renewal and stepwise progeny differentiation. In the Drosophila ovary, inner germarial sheath (IGS) cells form a niche for controlling germline stem cell (GSC) progeny differentiation. In this study, we have identified four IGS subpopulations, which form linearly arranged niche compartments for controlling GSC maintenance and multi-step progeny differentiation. Single-cell analysis of the adult ovary has identified four IGS subpopulations (IGS1-IGS4), the identities and cellular locations of which have been further confirmed by fluorescent in situ hybridization. IGS1 and IGS2 physically interact with GSCs and mitotic cysts to control GSC maintenance and cyst formation, respectively, whereas IGS3 and IGS4 physically interact with 16-cell cysts to regulate meiosis, oocyte development, and cyst morphological change. Finally, one follicle cell progenitor population has also been transcriptionally defined for facilitating future studies on follicle stem cell regulation. Therefore, this study has structurally revealed that the niche is organized into multiple compartments for orchestrating stepwise adult stem cell development and has also provided useful resources and tools for further functional characterization of the niche in the future.
Collapse
Affiliation(s)
- Renjun Tu
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Bo Duan
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Xiaoqing Song
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Shiyuan Chen
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Allison Scott
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Kate Hall
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Jillian Blanck
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Dustin DeGraffenreid
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Anoja Perera
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Jeff Haug
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Ting Xie
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
44
|
Juul NH, Stockman CA, Desai TJ. Niche Cells and Signals that Regulate Lung Alveolar Stem Cells In Vivo. Cold Spring Harb Perspect Biol 2020; 12:a035717. [PMID: 32179507 PMCID: PMC7706567 DOI: 10.1101/cshperspect.a035717] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The distal lung is a honeycomb-like collection of delicate gas exchange sacs called alveoli lined by two interspersed epithelial cell types: the cuboidal, surfactant-producing alveolar type II (AT2) and the flat, gas-exchanging alveolar type I (AT1) cell. During aging, a subset of AT2 cells expressing the canonical Wnt target gene, Axin2, function as stem cells, renewing themselves while generating new AT1 and AT2 cells. Wnt activity endows AT2 cells with proliferative competency, enabling them to respond to activating cues, and simultaneously blocks AT2 to AT1 cell transdifferentiation. Acute alveolar injury rapidly expands the AT2 stem cell pool by transiently inducing Wnt signaling activity in "bulk" AT2 cells, facilitating rapid epithelial repair. AT2 cell "stemness" is thus tightly regulated by access to Wnts, supplied by a specialized single-cell fibroblast niche during maintenance and by AT2 cells themselves during injury repair. Two non-AT2 "reserve" cell populations residing in the distal airways also contribute to alveolar repair, but only after widespread epithelial injury, when they rapidly proliferate, migrate, and differentiate into airway and alveolar lineages. Here, we review alveolar renewal and repair with a focus on the niches, rather than the stem cells, highlighting what is known about the cellular and molecular mechanisms by which they control stem cell activity in vivo.
Collapse
Affiliation(s)
- Nicholas H Juul
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Courtney A Stockman
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Tushar J Desai
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
45
|
Wang M, Chen X, Wu Y, Zheng Q, Chen W, Yan Y, Luan X, Shen C, Fang J, Zheng B, Yu J. RpS13 controls the homeostasis of germline stem cell niche through Rho1-mediated signals in the Drosophila testis. Cell Prolif 2020; 53:e12899. [PMID: 32896929 PMCID: PMC7574871 DOI: 10.1111/cpr.12899] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
Objectives Stem cell niche regulated the renewal and differentiation of germline stem cells (GSCs) in Drosophila. Previously, we and others identified a series of genes encoding ribosomal proteins that may contribute to the self‐renewal and differentiation of GSCs. However, the mechanisms that maintain and differentiate GSCs in their niches were not well understood. Materials and Methods Flies were used to generate tissue‐specific gene knockdown. Small interfering RNAs were used to knockdown genes in S2 cells. qRT‐PCR was used to examine the relative mRNA expression level. TUNEL staining or flow cytometry assays were used to detect cell survival. Immunofluorescence was used to determine protein localization and expression pattern. Results Herein, using a genetic manipulation approach, we investigated the role of ribosomal protein S13 (RpS13) in testes and S2 cells. We reported that RpS13 was required for the self‐renewal and differentiation of GSCs. We also demonstrated that RpS13 regulated cell proliferation and apoptosis. Mechanistically, we showed that RpS13 regulated the expression of ribosome subunits and could moderate the expression of the Rho1, DE‐cad and Arm proteins. Notably, Rho1 imitated the phenotype of RpS13 in both Drosophila testes and S2 cells, and recruited cell adhesions, which was mediated by the DE‐cad and Arm proteins. Conclusion These findings uncover a novel mechanism of RpS13 that mediates Rho1 signals in the stem cell niche of the Drosophila testis.
Collapse
Affiliation(s)
- Min Wang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Xia Chen
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Yibo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Qianwen Zheng
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Wanyin Chen
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Yidan Yan
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Xiaojin Luan
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jie Fang
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Bo Zheng
- State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jun Yu
- Department of Gynecology, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| |
Collapse
|
46
|
Ma B, Trieu TJ, Cheng J, Zhou S, Tang Q, Xie J, Liu JL, Zhao K, Habib SJ, Chen X. Differential Histone Distribution Patterns in Induced Asymmetrically Dividing Mouse Embryonic Stem Cells. Cell Rep 2020; 32:108003. [PMID: 32783931 PMCID: PMC7962874 DOI: 10.1016/j.celrep.2020.108003] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 11/03/2019] [Accepted: 07/15/2020] [Indexed: 12/14/2022] Open
Abstract
Wnt3a-coated beads can induce asymmetric divisions of mouse embryonic stem cells (mESCs), resulting in one self-renewed mESC and one differentiating epiblast stem cell. This provides an opportunity for studying histone inheritance pattern at a single-cell resolution in cell culture. Here, we report that mESCs with Wnt3a-bead induction display nonoverlapping preexisting (old) versus newly synthesized (new) histone H3 patterns, but mESCs without Wnt3a beads have largely overlapping patterns. Furthermore, H4K20me2/3, an old histone-enriched modification, displays a higher instance of asymmetric distribution on chromatin fibers from Wnt3a-induced mESCs than those from non-induced mESCs. These locally distinct distributions between old and new histones have both cellular specificity in Wnt3a-induced mESCs and molecular specificity for histones H3 and H4. Given that post-translational modifications at H3 and H4 carry the major histone modifications, our findings provide a mammalian cell culture system to study histone inheritance for maintaining stem cell fate and for resetting it during differentiation.
Collapse
Affiliation(s)
- Binbin Ma
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA; Research Center for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China; Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tung-Jui Trieu
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Ji Cheng
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
| | - Shuang Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Qingsong Tang
- Systems Biology Center, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, USA
| | - Jing Xie
- Research Center for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Ji-Long Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Keji Zhao
- Systems Biology Center, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, USA
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Xin Chen
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
47
|
DeLuca SZ, Ghildiyal M, Pang LY, Spradling AC. Differentiating Drosophila female germ cells initiate Polycomb silencing by regulating PRC2-interacting proteins. eLife 2020; 9:e56922. [PMID: 32773039 PMCID: PMC7438113 DOI: 10.7554/elife.56922] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/06/2020] [Indexed: 01/18/2023] Open
Abstract
Polycomb silencing represses gene expression and provides a molecular memory of chromatin state that is essential for animal development. We show that Drosophila female germline stem cells (GSCs) provide a powerful system for studying Polycomb silencing. GSCs have a non-canonical distribution of PRC2 activity and lack silenced chromatin like embryonic progenitors. As GSC daughters differentiate into nurse cells and oocytes, nurse cells, like embryonic somatic cells, silence genes in traditional Polycomb domains and in generally inactive chromatin. Developmentally controlled expression of two Polycomb repressive complex 2 (PRC2)-interacting proteins, Pcl and Scm, initiate silencing during differentiation. In GSCs, abundant Pcl inhibits PRC2-dependent silencing globally, while in nurse cells Pcl declines and newly induced Scm concentrates PRC2 activity on traditional Polycomb domains. Our results suggest that PRC2-dependent silencing is developmentally regulated by accessory proteins that either increase the concentration of PRC2 at target sites or inhibit the rate that PRC2 samples chromatin.
Collapse
Affiliation(s)
- Steven Z DeLuca
- Howard Hughes Medical Institute Research Laboratories Department of Embryology, Carnegie Institution for ScienceBaltimoreUnited States
| | - Megha Ghildiyal
- Howard Hughes Medical Institute Research Laboratories Department of Embryology, Carnegie Institution for ScienceBaltimoreUnited States
| | - Liang-Yu Pang
- Howard Hughes Medical Institute Research Laboratories Department of Embryology, Carnegie Institution for ScienceBaltimoreUnited States
| | - Allan C Spradling
- Howard Hughes Medical Institute Research Laboratories Department of Embryology, Carnegie Institution for ScienceBaltimoreUnited States
| |
Collapse
|
48
|
Amartuvshin O, Lin C, Hsu S, Kao S, Chen A, Tang W, Chou H, Chang D, Hsu Y, Hsiao B, Rastegari E, Lin K, Wang Y, Yao C, Chen G, Chen B, Hsu H. Aging shifts mitochondrial dynamics toward fission to promote germline stem cell loss. Aging Cell 2020; 19:e13191. [PMID: 32666649 PMCID: PMC7431834 DOI: 10.1111/acel.13191] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/20/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
Changes in mitochondrial dynamics (fusion and fission) are known to occur during stem cell differentiation; however, the role of this phenomenon in tissue aging remains unclear. Here, we report that mitochondrial dynamics are shifted toward fission during aging of Drosophila ovarian germline stem cells (GSCs), and this shift contributes to aging-related GSC loss. We found that as GSCs age, mitochondrial fragmentation and expression of the mitochondrial fission regulator, Dynamin-related protein (Drp1), are both increased, while mitochondrial membrane potential is reduced. Moreover, preventing mitochondrial fusion in GSCs results in highly fragmented depolarized mitochondria, decreased BMP stemness signaling, impaired fatty acid metabolism, and GSC loss. Conversely, forcing mitochondrial elongation promotes GSC attachment to the niche. Importantly, maintenance of aging GSCs can be enhanced by suppressing Drp1 expression to prevent mitochondrial fission or treating with rapamycin, which is known to promote autophagy via TOR inhibition. Overall, our results show that mitochondrial dynamics are altered during physiological aging, affecting stem cell homeostasis via coordinated changes in stemness signaling, niche contact, and cellular metabolism. Such effects may also be highly relevant to other stem cell types and aging-induced tissue degeneration.
Collapse
Affiliation(s)
- Oyundari Amartuvshin
- Molecular and Cell BiologyTaiwan International Graduate ProgramAcademia SinicaTaipeiTaiwan
- Graduate Institute of Life ScienceNational Defense Medical CenterTaipeiTaiwan
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
| | - Chi‐Hung Lin
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
| | - Shao‐Chun Hsu
- Imaging Core Facility at the Institute of Cellular and Organismic BiologyAcademia SinicaTaipeiTaiwan
| | - Shih‐Han Kao
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
- Present address:
Institute of ChemistryAcademia SinicaTaipeiTaiwan
| | - Alvin Chen
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
| | - Wei‐Chun Tang
- Research Center for Applied ScienceAcademia SinicaTaipeiTaiwan
| | - Han‐Lin Chou
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
| | - Dong‐Lin Chang
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
- The Affiliated Senior High School of National Taiwan Normal UniversityTaipeiTaiwan
| | - Yen‐Yang Hsu
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
- The Affiliated Senior High School of National Taiwan Normal UniversityTaipeiTaiwan
| | - Bai‐Shiou Hsiao
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
- The Affiliated Senior High School of National Taiwan Normal UniversityTaipeiTaiwan
| | | | - Kun‐Yang Lin
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
| | - Yu‐Ting Wang
- Molecular and Cell BiologyTaiwan International Graduate ProgramAcademia SinicaTaipeiTaiwan
- Graduate Institute of Life ScienceNational Defense Medical CenterTaipeiTaiwan
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
| | - Chi‐Kuang Yao
- Institute of Biological ChemistryAcademia SinicaTaipeiTaiwan
| | - Guang‐Chao Chen
- Institute of Biological ChemistryAcademia SinicaTaipeiTaiwan
| | - Bi‐Chang Chen
- Research Center for Applied ScienceAcademia SinicaTaipeiTaiwan
| | - Hwei‐Jan Hsu
- Molecular and Cell BiologyTaiwan International Graduate ProgramAcademia SinicaTaipeiTaiwan
- Graduate Institute of Life ScienceNational Defense Medical CenterTaipeiTaiwan
- Institute of Cellular and Organismic BiologyTaipeiTaiwan
| |
Collapse
|
49
|
Cordeiro IR, Tanaka M. Environmental Oxygen is a Key Modulator of Development and Evolution: From Molecules to Ecology. Bioessays 2020; 42:e2000025. [DOI: 10.1002/bies.202000025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/09/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Ingrid Rosenburg Cordeiro
- Department of Life Science and Technology Tokyo Institute of Technology B‐17, 4259 Nagatsuta‐cho, Midori‐ku Yokohama 226‐8501 Japan
| | - Mikiko Tanaka
- Department of Life Science and Technology Tokyo Institute of Technology B‐17, 4259 Nagatsuta‐cho, Midori‐ku Yokohama 226‐8501 Japan
| |
Collapse
|
50
|
Abstract
Stem cells including cancer stem cells (CSC) divide symmetrically or asymmetrically. Usually symmetric cell division makes two daughter cells of the same fate, either as stem cells or more differentiated progenies; while asymmetric cell division (ACD) produces daughter cells of different fates. In this review, we first provide an overview of ACD, and then discuss more molecular details of ACD using the well-characterized Drosophila neuroblast system as an example. Aiming to explore the connections between cell heterogeneity in cancers and the critical need of ACD for self-renewal and generating cell diversity, we then examine how cell division symmetry control impacts common features associated with CSCs, including niche competition, cancer dormancy, drug resistance, epithelial-mesenchymal transition (EMT) and its reverse process mesenchymal-epithelial transition (MET), and cancer stem cell plasticity. As CSC may underlie resistance to therapy and cancer metastasis, understanding how cell division mode is selected and executed in these cells will provide possible strategies to target CSC.
Collapse
Affiliation(s)
- Sreemita Majumdar
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Song-Tao Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| |
Collapse
|