1
|
Hodge RA, Bach EA. Mechanisms of Germline Stem Cell Competition across Species. Life (Basel) 2024; 14:1251. [PMID: 39459551 PMCID: PMC11509876 DOI: 10.3390/life14101251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
In this review, we introduce the concept of cell competition, which occurs between heterogeneous neighboring cell populations. Cells with higher relative fitness become "winners" that outcompete cells of lower relative fitness ("losers"). We discuss the idea of super-competitors, mutant cells that expand at the expense of wild-type cells. Work on adult stem cells (ASCs) has revealed principles of neutral competition, wherein ASCs can be stochastically lost and replaced, and of biased competition, in which a winning ASC with a competitive advantage replaces its neighbors. Germline stem cells (GSCs) are ASCs that are uniquely endowed with the ability to produce gametes and, therefore, impact the next generation. Mechanisms of GSC competition have been elucidated by studies in Drosophila gonads, tunicates, and the mammalian testis. Competition between ASCs is thought to underlie various forms of cancer, including spermatocytic tumors in the human testis. Paternal age effect (PAE) disorders are caused by de novo mutations in human GSCs that increase their competitive ability and make them more likely to be inherited, leading to skeletal and craniofacial abnormalities in offspring. Given its widespread effects on human health, it is important to study GSC competition to elucidate how cells can become winners or losers.
Collapse
Affiliation(s)
| | - Erika A. Bach
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA;
| |
Collapse
|
2
|
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
|
3
|
Melamed D, Choi A, Reilein A, Tavaré S, Kalderon D. Spatial regulation of Drosophila ovarian Follicle Stem Cell division rates and cell cycle transitions. PLoS Genet 2023; 19:e1010965. [PMID: 37747936 PMCID: PMC10553835 DOI: 10.1371/journal.pgen.1010965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/05/2023] [Accepted: 09/11/2023] [Indexed: 09/27/2023] Open
Abstract
Drosophila ovarian Follicle Stem Cells (FSCs) present a favorable paradigm for understanding how stem cell division and differentiation are balanced in communities where those activities are independent. FSCs also allow exploration of how this balance is integrated with spatial stem cell heterogeneity. Posterior FSCs become proliferative Follicle Cells (FCs), while anterior FSCs become quiescent Escort Cells (ECs) at about one fourth the frequency. A single stem cell can nevertheless produce both FCs and ECs because it can move between anterior and posterior locations. Studies based on EdU incorporation to approximate division rates suggested that posterior FSCs divide faster than anterior FSCs. However, direct measures of cell cycle times are required to ascertain whether FC output requires a net flow of FSCs from anterior to posterior. Here, by using live imaging and FUCCI cell-cycle reporters, we measured absolute division rates. We found that posterior FSCs cycle more than three times faster than anterior FSCs and produced sufficient new cells to match FC production. H2B-RFP dilution studies supported different cycling rates according to A/P location and facilitated live imaging, showing A/P exchange of FSCs in both directions, consistent with the dynamic equilibrium inferred from division rate measurements. Inversely graded Wnt and JAK-STAT pathway signals regulate FSC differentiation to ECs and FCs. JAK-STAT promotes both differentiation to FCs and FSC cycling, affording some coordination of these activities. When JAK-STAT signaling was manipulated to be spatially uniform, the ratio of posterior to anterior division rates was reduced but remained substantial, showing that graded JAK-STAT signaling only partly explains the graded cycling of FSCs. By using FUCCI markers, we found a prominent G2/M cycling restriction of posterior FSCs together with an A/P graded G1/S restriction, that JAK-STAT signaling promotes both G1/S and G2/M transitions, and that PI3 kinase signaling principally stimulates the G2/M transition.
Collapse
Affiliation(s)
- David Melamed
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
| | - Aaron Choi
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
| | - Amy Reilein
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
| | - Simon Tavaré
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
- Irving Institute for Cancer Dynamics & Department of Statistics, Columbia University, New York, New York State, United States of America
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, New York State, United States of America
| |
Collapse
|
4
|
Dong Z, Pang L, Liu Z, Sheng Y, Li X, Thibault X, Reilein A, Kalderon D, Huang J. Single-cell expression profile of Drosophila ovarian follicle stem cells illuminates spatial differentiation in the germarium. BMC Biol 2023; 21:143. [PMID: 37340484 PMCID: PMC10283321 DOI: 10.1186/s12915-023-01636-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND How stem cell populations are organized and regulated within adult tissues is important for understanding cancer origins and for developing cell replacement strategies. Paradigms such as mammalian gut stem cells and Drosophila ovarian follicle stem cells (FSC) are characterized by population asymmetry, in which stem cell division and differentiation are separately regulated processes. These stem cells behave stochastically regarding their contributions to derivative cells and also exhibit dynamic spatial heterogeneity. Drosophila FSCs provide an excellent model for understanding how a community of active stem cells maintained by population asymmetry is regulated. Here, we use single-cell RNA sequencing to profile the gene expression patterns of FSCs and their immediate derivatives to investigate heterogeneity within the stem cell population and changes associated with differentiation. RESULTS We describe single-cell RNA sequencing studies of a pre-sorted population of cells that include FSCs and the neighboring cell types, escort cells (ECs) and follicle cells (FCs), which they support. Cell-type assignment relies on anterior-posterior (AP) location within the germarium. We clarify the previously determined location of FSCs and use spatially targeted lineage studies as further confirmation. The scRNA profiles among four clusters are consistent with an AP progression from anterior ECs through posterior ECs and then FSCs, to early FCs. The relative proportion of EC and FSC clusters are in good agreement with the prevalence of those cell types in a germarium. Several genes with graded profiles from ECs to FCs are highlighted as candidate effectors of the inverse gradients of the two principal signaling pathways, Wnt and JAK-STAT, that guide FSC differentiation and division. CONCLUSIONS Our data establishes an important resource of scRNA-seq profiles for FSCs and their immediate derivatives that is based on precise spatial location and functionally established stem cell identity, and facilitates future genetic investigation of regulatory interactions guiding FSC behavior.
Collapse
Affiliation(s)
- Zhi Dong
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Lan Pang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Zhiguo Liu
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Yifeng Sheng
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China
| | - Xavier Thibault
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Amy Reilein
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| | - Jianhua Huang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
5
|
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
|
6
|
Abstract
A simple, universal and fundamental definition of adult stem cell communities is proposed. Key principles of cell lineage methods for defining adult stem cell numbers, locations and behaviors are critically evaluated, emphasizing the imperatives of capturing the full spectrum of individual stem cell behaviors, examining a variety of experimental time periods and avoiding unwarranted assumptions. The focus is first on defining fundamentals and then addresses stem cell heterogeneity, potential hierarchies and how individual cells serve the function of a stem cell community.
Collapse
|
7
|
Tatapudy S, Peralta J, Nystul T. Distinct roles of Bendless in regulating FSC niche competition and daughter cell differentiation. Development 2021; 148:dev199630. [PMID: 35020878 PMCID: PMC8645206 DOI: 10.1242/dev.199630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/13/2021] [Indexed: 04/05/2024]
Abstract
A major goal in the study of adult stem cells is to understand how cell fates are specified at the proper time and place to facilitate tissue homeostasis. Here, we found that an E2 ubiquitin ligase, Bendless (Ben), has multiple roles in the Drosophila ovarian epithelial follicle stem cell (FSC) lineage. First, Ben is part of the JNK signaling pathway, and we found that it, as well as other JNK pathway genes, are essential for differentiation of FSC daughter cells. Our data suggest that JNK signaling promotes differentiation by suppressing the activation of the EGFR effector, ERK. Also, we found that loss of ben, but not the JNK kinase hemipterous, resulted in an upregulation of hedgehog signaling, increased proliferation and increased niche competition. Lastly, we demonstrate that the hypercompetition phenotype caused by loss of ben is suppressed by decreasing the rate of proliferation or knockdown of the hedgehog pathway effector, Smoothened (Smo). Taken together, our findings reveal a new layer of regulation in which a single gene influences cell signaling at multiple stages of differentiation in the early FSC lineage.
Collapse
Affiliation(s)
| | | | - Todd Nystul
- Department of Anatomy and Department of OB/Gyn-RS, University of California, San Francisco, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
8
|
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: 10] [Impact Index Per Article: 2.5] [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
|
9
|
Slaidina M, Gupta S, Banisch TU, Lehmann R. A single-cell atlas reveals unanticipated cell type complexity in Drosophila ovaries. Genome Res 2021; 31:1938-1951. [PMID: 34389661 DOI: 10.1101/gr.274340.120] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 08/09/2021] [Indexed: 11/24/2022]
Abstract
Organ function relies on the spatial organization and functional coordination of numerous cell types. The Drosophila ovary is a widely used model system to study the cellular activities underlying organ function, including stem cell regulation, cell signaling and epithelial morphogenesis. However, the relative paucity of cell type-specific reagents hinders investigation of molecular functions at the appropriate cellular resolution. Here, we used single-cell RNA sequencing to characterize all cell types of the stem cell compartment and early follicles of the Drosophila ovary. We computed transcriptional signatures and identified specific markers for nine states of germ cell differentiation, and 23 somatic cell types and subtypes. We uncovered an unanticipated diversity of escort cells, the somatic cells that directly interact with differentiating germline cysts. Three escort cell subtypes reside in discrete anatomical positions, and express distinct sets of secreted and transmembrane proteins, suggesting that diverse micro-environments support the progressive differentiation of germ cells. Finally, we identified 17 follicle cell subtypes, and characterized their transcriptional profiles. Altogether, we provide a comprehensive resource of gene expression, cell type-specific markers, spatial coordinates and functional predictions for 34 ovarian cell types and subtypes.
Collapse
Affiliation(s)
| | - Selena Gupta
- Skirball Institute, NYU Grossman School of Medicine
| | | | | |
Collapse
|
10
|
Marca JEL, Somers WG. The Drosophila gonads: models for stem cell proliferation, self-renewal, and differentiation. AIMS GENETICS 2021. [DOI: 10.3934/genet.2014.1.55] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AbstractThe male and female gonads of Drosophila melanogaster have developed into powerful model systems for both the study of stem cell behaviours, and for understanding how stem cell misregulation can lead to cancers. Using these systems, one is able to observe and manipulate the resident stem cell populations in vivo with a great deal of licence. The tractability of the testis and ovary also allow researchers to explore a range of cellular mechanisms, such as proliferation and polarity, as well as the influence exerted by the local environment through a host of highly-conserved signalling pathways. Importantly, many of the cellular behaviours and processes studied in the Drosophila testis and ovary are known to be disrupted, or otherwise misregulated, in human tumourigenic cells. Here, we review the mechanisms relating to stem cell behaviour, though we acknowledge there are many other fascinating aspects of gametogenesis, including the invasive behaviour of migratory border cells in the Drosophila ovary that, though relevant to the study of tumourigenesis, will unfortunately not be covered.
Collapse
Affiliation(s)
- John E. La Marca
- Department of Genetics, La Trobe University, Melbourne, VIC 3086, Australia
| | | |
Collapse
|
11
|
Grmai L, Harsh S, Lu S, Korman A, Deb IB, Bach EA. Transcriptomic analysis of feminizing somatic stem cells in the Drosophila testis reveals putative downstream effectors of the transcription factor Chinmo. G3 (BETHESDA, MD.) 2021; 11:jkab067. [PMID: 33751104 PMCID: PMC8759813 DOI: 10.1093/g3journal/jkab067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/24/2021] [Indexed: 11/12/2022]
Abstract
One of the best examples of sexual dimorphism is the development and function of the gonads, ovaries and testes, which produce sex-specific gametes, oocytes, and spermatids, respectively. The development of these specialized germ cells requires sex-matched somatic support cells. The sexual identity of somatic gonadal cells is specified during development and must be actively maintained during adulthood. We previously showed that the transcription factor Chinmo is required to ensure the male sexual identity of somatic support cells in the Drosophila melanogaster testis. Loss of chinmo from male somatic gonadal cells results in feminization: they transform from squamous to epithelial-like cells that resemble somatic cells in the female gonad but fail to properly ensheath the male germline, causing infertility. To identify potential target genes of Chinmo, we purified somatic cells deficient for chinmo from the adult Drosophila testis and performed next-generation sequencing to compare their transcriptome to that of control somatic cells. Bioinformatics revealed 304 and 1549 differentially upregulated and downregulated genes, respectively, upon loss of chinmo in early somatic cells. Using a combination of methods, we validated several differentially expressed genes. These data sets will be useful resources to the community.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Sneh Harsh
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Sean Lu
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Aryeh Korman
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Ishan B Deb
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Erika A Bach
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
12
|
A Progressive Somatic Cell Niche Regulates Germline Cyst Differentiation in the Drosophila Ovary. Curr Biol 2021; 31:840-852.e5. [PMID: 33340458 DOI: 10.1016/j.cub.2020.11.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 10/02/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022]
Abstract
In the germarium of the Drosophila ovary, developing germline cysts are surrounded by a population of somatic escort cells that are known to function as the niche cells for germline differentiation;1 however, the underlying molecular mechanisms of this niche function remain poorly understood. Through single-cell gene expression profiling combined with genetic analyses, we here demonstrate that the escort cells can be spatially and functionally divided into two successive domains. The anterior escort cells (aECs) specifically produce ecdysone, which acts on the cystoblast to promote synchronous cell division, whereas the posterior escort cells (pECs) respond to ecdysone signaling and regulate soma-germline cell adhesion to promote the transition from 16-cell cyst-to-egg chamber formation. The patterning of the aEC and pEC domains is independent of the germline but is dependent on JAK/STAT signaling activity, which emanates from the posterior. Thus, a heterogeneous population of escort cells constitutes a stepwise niche environment to orchestrate cystoblast division and differentiation toward egg chamber formation.
Collapse
|
13
|
Melamed D, Kalderon D. Opposing JAK-STAT and Wnt signaling gradients define a stem cell domain by regulating differentiation at two borders. eLife 2020; 9:61204. [PMID: 33135631 PMCID: PMC7695452 DOI: 10.7554/elife.61204] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Many adult stem cell communities are maintained by population asymmetry, where stochastic behaviors of multiple individual cells collectively result in a balance between stem cell division and differentiation. We investigated how this is achieved for Drosophila Follicle Stem Cells (FSCs) by spatially-restricted niche signals. FSCs produce transit-amplifying Follicle Cells (FCs) from their posterior face and quiescent Escort Cells (ECs) to their anterior. We show that JAK-STAT pathway activity, which declines from posterior to anterior, dictates the pattern of divisions over the FSC domain, promotes more posterior FSC locations and conversion to FCs, while opposing EC production. Wnt pathway activity declines from the anterior, promotes anterior FSC locations and EC production, and opposes FC production. The pathways combine to define a stem cell domain through concerted effects on FSC differentiation to ECs and FCs at either end of opposing signaling gradients, and impose a pattern of proliferation that matches derivative production.
Collapse
Affiliation(s)
- David Melamed
- Department of Biological Sciences, Columbia University, New York, United States
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, United States
| |
Collapse
|
14
|
Waghmare I, Wang X, Page-McCaw A. Dally-like protein sequesters multiple Wnt ligands in the Drosophila germarium. Dev Biol 2020; 464:88-102. [PMID: 32473955 DOI: 10.1016/j.ydbio.2020.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/02/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
Cells in multicellular organisms rely on secreted ligands for development and morphogenesis. Several mechanisms modulate the availability and distribution of secreted ligands, determining their ability to signal locally and at long range from their source. One of these mechanisms is Dally-like protein (Dlp), a cell-surface glypican that exhibits biphasic functions in Drosophila wing discs, promoting Wg signaling at long-range from Wg source cells and inhibiting Wg signaling near source cells. In the germarium at the tip of the ovary, Dlp promotes long-range distribution of Wg from cap cells to follicle stem cells. However, the germarium also expresses other Wnts - Wnt2, Wnt4, and Wnt6 - that function locally in escort cells to promote oogenesis. Whether and how local functions of these Wnts are regulated remains unknown. Here we show that the dlp overexpression phenotype is multifaceted and phenocopies multiple Wnt loss-of-function phenotypes. Each aspect of dlp overexpression phenotype is suppressed by co-expression of individual Wnts, and the suppression pattern exhibited by each Wnt suggests that Wnts have functional specificity in the germarium. Further, dlp knockdown phenocopies Wnt gain-of-function phenotypes. Together these data show that Dlp inhibits the functions of each Wnt. All four Wnts co-immunoprecipitate with Dlp in S2R+ cells, suggesting that in the germarium, Dlp sequesters Wnts to inhibit local paracrine Wnt signaling. Our results indicate that Dlp modulates the availability of multiple extracellular Wnts for local paracrine Wnt signaling in the germarium.
Collapse
Affiliation(s)
- Indrayani Waghmare
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| | - Xiaoxi Wang
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| |
Collapse
|
15
|
Jevitt A, Chatterjee D, Xie G, Wang XF, Otwell T, Huang YC, Deng WM. A single-cell atlas of adult Drosophila ovary identifies transcriptional programs and somatic cell lineage regulating oogenesis. PLoS Biol 2020; 18:e3000538. [PMID: 32339165 PMCID: PMC7205450 DOI: 10.1371/journal.pbio.3000538] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 05/07/2020] [Accepted: 03/27/2020] [Indexed: 12/28/2022] Open
Abstract
Oogenesis is a complex developmental process that involves spatiotemporally regulated coordination between the germline and supporting, somatic cell populations. This process has been modeled extensively using the Drosophila ovary. Although different ovarian cell types have been identified through traditional means, the large-scale expression profiles underlying each cell type remain unknown. Using single-cell RNA sequencing technology, we have built a transcriptomic data set for the adult Drosophila ovary and connected tissues. Using this data set, we identified the transcriptional trajectory of the entire follicle-cell population over the course of their development from stem cells to the oogenesis-to-ovulation transition. We further identify expression patterns during essential developmental events that take place in somatic and germline cell types such as differentiation, cell-cycle switching, migration, symmetry breaking, nurse-cell engulfment, egg-shell formation, and corpus luteum signaling. Extensive experimental validation of unique expression patterns in both ovarian and nearby, nonovarian cells also led to the identification of many new cell type-and stage-specific markers. The inclusion of several nearby tissue types in this data set also led to our identification of functional convergence in expression between distantly related cell types such as the immune-related genes that were similarly expressed in immune cells (hemocytes) and ovarian somatic cells (stretched cells) during their brief phagocytic role in nurse-cell engulfment. Taken together, these findings provide new insight into the temporal regulation of genes in a cell-type specific manner during oogenesis and begin to reveal the relatedness in expression between cell and tissues types.
Collapse
Affiliation(s)
- Allison Jevitt
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Deeptiman Chatterjee
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Gengqiang Xie
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Xian-Feng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Taylor Otwell
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
16
|
Rust K, Nystul T. Signal transduction in the early Drosophila follicle stem cell lineage. CURRENT OPINION IN INSECT SCIENCE 2020; 37:39-48. [PMID: 32087562 PMCID: PMC7155752 DOI: 10.1016/j.cois.2019.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 05/08/2023]
Abstract
The follicle stem cell (FSC) lineage in the Drosophila ovary is a highly informative model of in vivo epithelial stem cell biology. Studies over the past 30 years have identified roles for every major signaling pathway in the early FSC lineage. These pathways regulate a wide variety of cell behaviors, including self-renewal, proliferation, survival and differentiation. Studies of cell signaling in the follicle epithelium have provided new insights into how these cell behaviors are coordinated within an epithelial stem cell lineage and how signaling pathways interact with each other in the native, in vivo context of a living tissue. Here, we review these studies, with a particular focus on how these pathways specify differences between the FSCs and their daughter cells. We also describe common themes that have emerged from these studies, and highlight new research directions that have been made possible by the detailed understanding of the follicle epithelium.
Collapse
|
17
|
Hayashi Y, Yoshinari Y, Kobayashi S, Niwa R. The regulation of Drosophila ovarian stem cell niches by signaling crosstalk. CURRENT OPINION IN INSECT SCIENCE 2020; 37:23-29. [PMID: 32087560 DOI: 10.1016/j.cois.2019.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/06/2019] [Accepted: 10/30/2019] [Indexed: 06/10/2023]
Abstract
The Drosophila female ovary is an excellent model for investigating how multiple stem cell types are coordinately regulated in vivo. The ovary contains at least two stem cell types, germline stem cells (GSCs) and somatic follicular stem cells (FSCs). Although GSCs and FSCs are maintained within a distinct extra-cellular microenvironment, known as a niche, they share some common signaling molecules to generate their own niche. To properly maintain these stem cell types, understanding how signaling molecules are regulated is essential. In this review, we summarize the recent understanding of the mechanisms maintaining GSCs and FSCs from the perspective of growth factor regulation and discuss how these regulatory mechanisms contribute to stem cell maintenance, competition, and survival.
Collapse
Affiliation(s)
- Yoshiki Hayashi
- Life Science Center for Survival Dynamics (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
| | - Yuto Yoshinari
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Satoru Kobayashi
- Life Science Center for Survival Dynamics (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
18
|
Niwa R, Kai T. Editorial overview: Stem cells orchestrate oogenesis: a lesson from the fruit fly, Drosophila melanogaster. CURRENT OPINION IN INSECT SCIENCE 2020; 37:iii-v. [PMID: 32199590 DOI: 10.1016/j.cois.2020.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Affiliation(s)
- Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki 305-8577, Japan.
| | - Toshie Kai
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
19
|
Herrera SC, Bach EA. JAK/STAT signaling in stem cells and regeneration: from Drosophila to vertebrates. Development 2019; 146:dev167643. [PMID: 30696713 PMCID: PMC6361132 DOI: 10.1242/dev.167643] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
The JAK/STAT pathway is a conserved metazoan signaling system that transduces cues from extracellular cytokines into transcriptional changes in the nucleus. JAK/STAT signaling is best known for its roles in immunity. However, recent work has demonstrated that it also regulates critical homeostatic processes in germline and somatic stem cells, as well as regenerative processes in several tissues, including the gonad, intestine and appendages. Here, we provide an overview of JAK/STAT signaling in stem cells and regeneration, focusing on Drosophila and highlighting JAK/STAT pathway functions in proliferation, survival and cell competition that are conserved between Drosophila and vertebrates.
Collapse
Affiliation(s)
- Salvador C Herrera
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Erika A Bach
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
- Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
20
|
Drosophila Glypicans Regulate Follicle Stem Cell Maintenance and Niche Competition. Genetics 2018; 209:537-549. [PMID: 29632032 DOI: 10.1534/genetics.118.300839] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/04/2018] [Indexed: 01/06/2023] Open
Abstract
Adult stem cells reside in specialized microenvironments called niches, which provide signals for stem cells to maintain their undifferentiated and self-renewing state. To maintain stem cell quality, several types of stem cells are known to be regularly replaced by progenitor cells through niche competition. However, the cellular and molecular bases for stem cell competition for niche occupancy are largely unknown. Here, we show that two Drosophila members of the glypican family of heparan sulfate proteoglycans (HSPGs), Dally and Dally-like (Dlp), differentially regulate follicle stem cell (FSC) maintenance and competitiveness for niche occupancy. Lineage analyses of glypican mutant FSC clones showed that dally is essential for normal FSC maintenance. In contrast, dlp is a hypercompetitive mutation: dlp mutant FSC progenitors often eventually occupy the entire epithelial sheet. RNA interference knockdown experiments showed that Dally and Dlp play both partially redundant and distinct roles in regulating Jak/Stat, Wg, and Hh signaling in FSCs. The Drosophila FSC system offers a powerful genetic model to study the mechanisms by which HSPGs exert specific functions in stem cell replacement and competition.
Collapse
|
21
|
Division-independent differentiation mandates proliferative competition among stem cells. Proc Natl Acad Sci U S A 2018; 115:E3182-E3191. [PMID: 29555768 DOI: 10.1073/pnas.1718646115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cancer-initiating gatekeeper mutations that arise in stem cells would be especially potent if they stabilize and expand an affected stem cell lineage. It is therefore important to understand how different stem cell organization strategies promote or prevent variant stem cell amplification in response to different types of mutation, including those that activate proliferation. Stem cell numbers can be maintained constant while producing differentiated products through individually asymmetrical division outcomes or by population asymmetry strategies in which individual stem cell lineages necessarily compete for niche space. We considered alternative mechanisms underlying population asymmetry and used quantitative modeling to predict starkly different consequences of altering proliferation rate: A variant, faster proliferating mutant stem cell should compete better only when stem cell division and differentiation are independent processes. For most types of stem cells, it has not been possible to ascertain experimentally whether division and differentiation are coupled. However, Drosophila follicle stem cells (FSCs) provided a favorable system with which to investigate population asymmetry mechanisms and also for measuring the impact of altered proliferation on competition. We found from detailed cell lineage studies that division and differentiation of an individual FSC are not coupled. We also found that FSC representation, reflecting maintenance and amplification, was highly responsive to genetic changes that altered only the rate of FSC proliferation. The FSC paradigm therefore provides definitive experimental evidence for the general principle that relative proliferation rate will always be a major determinant of competition among stem cells specifically when stem cell division and differentiation are independent.
Collapse
|
22
|
Rossi L, Salvetti A. Planarian stem cell niche, the challenge for understanding tissue regeneration. Semin Cell Dev Biol 2018. [PMID: 29534938 DOI: 10.1016/j.semcdb.2018.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Stem cell fate depends on surrounding microenvironment, the so called niche. For this reason, understanding stem cell niche is one of the most challenging target in cell biology field and need to be unraveled with in vivo studies. Planarians offer this unique opportunity, as their stem cells, the neoblasts, are abundant, highly characterized and genetically modifiable by RNA interference in alive animals. However, despite impressive advances have been done in the understanding planarian stem cells and regeneration, only a few information is available in defining signals from differentiated tissues, which affect neoblast stemness and fate. Here, we review on molecular factors that have been found activated in differentiated tissues and directly or indirectly affect neoblast behavior, and we suggest future directions for unravelling this challenge in understanding planarian stem cells.
Collapse
Affiliation(s)
- Leonardo Rossi
- Departement of Clinical and Experimental Medicine, Unit of Experimental Biology and Genetics, University of Pisa, Via Volta 4 Pisa, Italy
| | - Alessandra Salvetti
- Departement of Clinical and Experimental Medicine, Unit of Experimental Biology and Genetics, University of Pisa, Via Volta 4 Pisa, Italy.
| |
Collapse
|
23
|
Waghmare I, Page-McCaw A. Wnt Signaling in Stem Cell Maintenance and Differentiation in the Drosophila Germarium. Genes (Basel) 2018; 9:genes9030127. [PMID: 29495453 PMCID: PMC5867848 DOI: 10.3390/genes9030127] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 01/19/2023] Open
Abstract
Wnt signaling is a conserved regulator of stem cell behaviors, and the Drosophila germarium has been an important model tissue for the study of stem cell maintenance, differentiation, and proliferation. Here we review Wnt signaling in the germarium, which houses two distinct types of ovarian stem cells: the anteriorly located germline stem cells (GSCs), which give rise to oocytes; and the mid-posteriorly located follicle stem cells (FSCs), which give rise to the somatic follicle cells that cover a developing oocyte. The maintenance and proliferation of GSCs and FSCs is regulated by the stem cell niches, whereas differentiation of the germline is regulated by the differentiation niche. Four distinct Wnt ligands are localized in the germarium, and we focus review on how these Wnt ligands and Wnt signaling affects maintenance and differentiation of both germline and follicle stem cells in their respective niches.
Collapse
Affiliation(s)
- Indrayani Waghmare
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA.
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA.
| |
Collapse
|
24
|
Wang X, Page-McCaw A. Wnt6 maintains anterior escort cells as an integral component of the germline stem cell niche. Development 2018; 145:dev.158527. [PMID: 29361569 PMCID: PMC5818006 DOI: 10.1242/dev.158527] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 12/22/2017] [Indexed: 12/27/2022]
Abstract
Stem cells reside in a niche, a local environment whose cellular and molecular complexity is still being elucidated. In Drosophila ovaries, germline stem cells depend on cap cells for self-renewing signals and physical attachment. Germline stem cells also contact the anterior escort cells, and here we report that anterior escort cells are absolutely required for germline stem cell maintenance. When escort cells die from impaired Wnt signaling or hid expression, the loss of anterior escort cells causes loss of germline stem cells. Anterior escort cells function as an integral niche component by promoting DE-cadherin anchorage and by transiently expressing the Dpp ligand to promote full-strength BMP signaling in germline stem cells. Anterior escort cells are maintained by Wnt6 ligands produced by cap cells; without Wnt6 signaling, anterior escort cells die leaving vacancies in the niche, leading to loss of germline stem cells. Our data identify anterior escort cells as constituents of the germline stem cell niche, maintained by a cap cell-produced Wnt6 survival signal.
Collapse
Affiliation(s)
- Xiaoxi Wang
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
25
|
Dai W, Peterson A, Kenney T, Burrous H, Montell DJ. Quantitative microscopy of the Drosophila ovary shows multiple niche signals specify progenitor cell fate. Nat Commun 2017; 8:1244. [PMID: 29093440 PMCID: PMC5665863 DOI: 10.1038/s41467-017-01322-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 09/09/2017] [Indexed: 01/28/2023] Open
Abstract
Adult stem cells commonly give rise to transit-amplifying progenitors, whose progeny differentiate into distinct cell types. It is unclear if stem cell niche signals coordinate fate decisions within the progenitor pool. Here we use quantitative analysis of Wnt, Hh, and Notch signalling reporters and the cell fate markers Eyes Absent (Eya) and Castor (Cas) to study the effects of hyper-activation and loss of niche signals on progenitor development in the Drosophila ovary. Follicle stem cell (FSC) progeny adopt distinct polar, stalk, and main body cell fates. We show that Wnt signalling transiently inhibits expression of the main body cell fate determinant Eya, and Wnt hyperactivity strongly biases cells towards polar and stalk fates. Hh signalling independently controls the proliferation to differentiation transition. Notch is permissive but not instructive for differentiation of multiple cell types. These findings reveal that multiple niche signals coordinate cell fates and differentiation of progenitor cells.
Collapse
Affiliation(s)
- Wei Dai
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA
| | - Amy Peterson
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA
| | - Thomas Kenney
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA
| | - Haley Burrous
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA
| | - Denise J Montell
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
26
|
Cook MS, Cazin C, Amoyel M, Yamamoto S, Bach E, Nystul T. Neutral Competition for Drosophila Follicle and Cyst Stem Cell Niches Requires Vesicle Trafficking Genes. Genetics 2017; 206:1417-1428. [PMID: 28512187 PMCID: PMC5500140 DOI: 10.1534/genetics.117.201202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/10/2017] [Indexed: 12/03/2022] Open
Abstract
The process of selecting for cellular fitness through competition plays a critical role in both development and disease. The germarium, a structure at the tip of the ovariole of a Drosophila ovary, contains two follicle stem cells (FSCs) that undergo neutral competition for the stem cell niche. Using the FSCs as a model, we performed a genetic screen through a collection of 126 mutants in essential genes on the X chromosome to identify candidates that increase or decrease competition for the FSC niche. We identified ∼55 and 6% of the mutations screened as putative FSC hypo- or hyper-competitors, respectively. We found that a large majority of mutations in vesicle trafficking genes (11 out of the 13 in the collection of mutants) are candidate hypo-competition alleles, and we confirmed the hypo-competition phenotype for four of these alleles. We also show that Sec16 and another COPII vesicle trafficking component, Sar1, are required for follicle cell differentiation. Lastly, we demonstrate that, although some components of vesicle trafficking are also required for neutral competition in the cyst stem cells of the testis, there are important tissue-specific differences. Our results demonstrate a critical role for vesicle trafficking in stem cell niche competition and differentiation, and we identify a number of putative candidates for further exploration.
Collapse
Affiliation(s)
- Matthew S Cook
- Center for Reproductive Sciences, University of California, San Francisco, California 94143-0452
- Department of Anatomy, University of California, San Francisco, California 94143-0452
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, California 94143-0452
| | - Coralie Cazin
- The Helen and Martin Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York 10016
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York 10016
| | - Marc Amoyel
- The Helen and Martin Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York 10016
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York 10016
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, United Kingdom
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
| | - Erika Bach
- The Helen and Martin Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York 10016
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York 10016
| | - Todd Nystul
- Center for Reproductive Sciences, University of California, San Francisco, California 94143-0452
- Department of Anatomy, University of California, San Francisco, California 94143-0452
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, California 94143-0452
| |
Collapse
|
27
|
The Hippo pathway acts downstream of the Hedgehog signaling to regulate follicle stem cell maintenance in the Drosophila ovary. Sci Rep 2017; 7:4480. [PMID: 28667262 PMCID: PMC5493701 DOI: 10.1038/s41598-017-04052-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/09/2017] [Indexed: 11/18/2022] Open
Abstract
The Hippo pathway is conserved and plays important roles in organ size control. The core components of the Hippo pathway are two kinases Hippo (Hpo), Warts (Wts), and a transcription-co-activator Yorkie (Yki). Yki activity is regulated by phosphorylation, which affects its nuclear localization and stability. To determine the role of the Hippo pathway in stem cells, we examine follicle stem cells (FSCs) in the Drosophila ovary. Yki is detected in the nucleus of FSCs. Knockdown of yki in the follicle cell lineage leads to a disruption of the follicular epithelium. Mitotic clones of FSCs mutant for hpo or wts are maintained in the niche and tend to replace the other FSCs, and FSCs mutant for yki are rapidly lost, demonstrating that the Hippo pathway is both required and sufficient for FSC maintenance. Using genetic interaction analyses, we demonstrate that the Hedgehog pathway acts upstream of the Hippo pathway in regulating FSC maintenance. The nuclear localization of Yki is enhanced when the Hedgehog signaling is activated. Furthermore, a constitutively active but not a wild-type Yki promotes FSC maintenance as activation of the Hedgehog signaling does, suggesting that the Hedgehog pathway regulates Yki through a post-translational mechanism in maintaining FSCs.
Collapse
|
28
|
Huang J, Reilein A, Kalderon D. Yorkie and Hedgehog independently restrict BMP production in escort cells to permit germline differentiation in the Drosophila ovary. Development 2017; 144:2584-2594. [PMID: 28619819 DOI: 10.1242/dev.147702] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 06/08/2017] [Indexed: 12/13/2022]
Abstract
Multiple signaling pathways guide the behavior and differentiation of both germline stem cells (GSCs) and somatic follicle stem cells (FSCs) in the Drosophila germarium, necessitating careful control of signal generation, range and responses. Signal integration involves escort cells (ECs), which promote differentiation of the GSC derivatives they envelop, provide niche signals for FSCs and derive directly from FSCs in adults. Hedgehog (Hh) signaling induces the Hippo pathway effector Yorkie (Yki) to promote proliferation and maintenance of FSCs, but Hh also signals to ECs, which are quiescent. Here, we show that in ECs both Hh and Yki limit production of BMP ligands to allow germline differentiation. Loss of Yki produced a more severe germarial phenotype than loss of Hh signaling and principally induced a different BMP ligand. Moreover, Yki activity reporters and epistasis tests showed that Yki does not mediate the key actions of Hh signaling in ECs. Thus, both the coupling and output of the Hh and Yki signaling pathways differ between FSCs and ECs despite their proximity and the fact that FSCs give rise directly to ECs.
Collapse
Affiliation(s)
- Jianhua Huang
- State Key Laboratory of Rice Biology, Institute of Insect Sciences, Zhejiang University, Hangzhou, 310058, China.,Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave., New York, NY 10027, USA
| | - Amy Reilein
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave., New York, NY 10027, USA
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave., New York, NY 10027, USA
| |
Collapse
|
29
|
Reilein A, Melamed D, Park KS, Berg A, Cimetta E, Tandon N, Vunjak-Novakovic G, Finkelstein S, Kalderon D. Alternative direct stem cell derivatives defined by stem cell location and graded Wnt signalling. Nat Cell Biol 2017; 19:433-444. [PMID: 28414313 PMCID: PMC5672635 DOI: 10.1038/ncb3505] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 03/06/2017] [Indexed: 12/15/2022]
Abstract
Adult stem cells provide a renewable source of differentiated cells for a wide variety of tissues and generally give rise to multiple cell types. Basic principles of stem cell organization and regulation underlying this behavior are emerging. Local niche signals maintain stem cells, while different sets of signals act outside the niche to diversify initially equivalent stem cell progeny. Here we show that Drosophila ovarian Follicle Stem Cells (FSCs) produced two distinct cell types directly. This cell fate choice was determined by the A/P position of an FSC and by the magnitude of spatially graded Wnt pathway activity. These findings reveal a paradigm of immediate diversification of stem cell derivatives according to stem cell position within a larger population, guided by a graded niche signal. We also found that FSCs strongly resemble mammalian intestinal stem cells in many aspects of their organization, including population asymmetry and dynamic heterogeneity.
Collapse
Affiliation(s)
- Amy Reilein
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - David Melamed
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Karen Sophia Park
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Ari Berg
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Elisa Cimetta
- Department of Biomedical Engineering, Columbia University, New York, New York 10032, USA
| | - Nina Tandon
- Department of Biomedical Engineering, Columbia University, New York, New York 10032, USA
| | | | - Sarah Finkelstein
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
30
|
Riechmann V. In vivo RNAi in the Drosophila Follicular Epithelium: Analysis of Stem Cell Maintenance, Proliferation, and Differentiation. Methods Mol Biol 2017; 1622:185-206. [PMID: 28674810 DOI: 10.1007/978-1-4939-7108-4_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In vivo RNAi in Drosophila facilitates simple and rapid analysis of gene functions in a cell- or tissue-specific manner. The versatility of the UAS-GAL4 system allows to control exactly where and when during development the function of a gene is depleted. The epithelium of the ovary is a particularly good model to study in a living animal how stem cells are maintained and how their descendants proliferate and differentiate. Here I provide basic information about the publicly available reagents for in vivo RNAi, and I describe how the oogenesis system can be applied to analyze stem cells and epithelial development at a histological level. Moreover, I give helpful hints to optimize the use of the UAS-GAL4 system for RNAi induction in the follicular epithelium. Finally, I provide detailed step-by-step protocols for ovary dissection, antibody stainings, and ovary mounting for microscopic analysis.
Collapse
Affiliation(s)
- Veit Riechmann
- Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg University, Ludolf-Krehl-Strasse 13-17, D-68167, Mannheim, Germany.
| |
Collapse
|
31
|
Silva D, Jemc JC. Sorting Out Identities: An Educational Primer for Use with "Novel Tools for Genetic Manipulation of Follicle Stem Cells in the Drosophila Ovary Reveal an Integrin-Dependent Transition from Quiescence to Proliferation". Genetics 2015; 201:13-22. [PMID: 26354974 PMCID: PMC4566258 DOI: 10.1534/genetics.115.179911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Organisms are made up of thousands of different cell types that must migrate, proliferate, and interact with each other to yield functional organ systems and ultimately a viable organism. A characteristic that distinguishes one cell type from another is the set of genes that it expresses. An article by Hartman et al. in the April 2015 issue of GENETICS identified methods to uniquely identify different cell populations during oogenesis, providing valuable tools for future studies. This Primer article provides background information on the Drosophila ovary as a system in which to study stem cell regulation, mechanisms for regulating gene expression, and the techniques used by Hartman et al. to identify specific cell populations and study their function.
Collapse
Affiliation(s)
- Diane Silva
- Department of Biology, Loyola University Chicago, Chicago, Illinois 60660
| | - Jennifer C Jemc
- Department of Biology, Loyola University Chicago, Chicago, Illinois 60660
| |
Collapse
|
32
|
Hartman TR, Ventresca EM, Hopkins A, Zinshteyn D, Singh T, O'Brien JA, Neubert BC, Hartman MG, Schofield HK, Stavrides KP, Talbot DE, Riggs DJ, Pritchard C, O'Reilly AM. Novel tools for genetic manipulation of follicle stem cells in the Drosophila ovary reveal an integrin-dependent transition from quiescence to proliferation. Genetics 2015; 199:935-57. [PMID: 25680813 PMCID: PMC4391569 DOI: 10.1534/genetics.114.173617] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/09/2015] [Indexed: 01/11/2023] Open
Abstract
In many tissues, the presence of stem cells is inferred by the capacity of the tissue to maintain homeostasis and undergo repair after injury. Isolation of self-renewing cells with the ability to generate the full array of cells within a given tissue strongly supports this idea, but the identification and genetic manipulation of individual stem cells within their niche remain a challenge. Here we present novel methods for marking and genetically altering epithelial follicle stem cells (FSCs) within the Drosophila ovary. Using these new tools, we define a sequential multistep process that comprises transitioning of FSCs from quiescence to proliferation. We further demonstrate that integrins are cell-autonomously required within FSCs to provide directional signals that are necessary at each step of this process. These methods may be used to define precise roles for specific genes in the sequential events that occur during FSC division after a period of quiescence.
Collapse
Affiliation(s)
- Tiffiney R Hartman
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Erin M Ventresca
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Anthony Hopkins
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Daniel Zinshteyn
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Tanu Singh
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111 Molecular Cell Biology and Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Jenny A O'Brien
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111 Department of Cancer Biology and Genetics, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Benjamin C Neubert
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111 North Penn High School, Lansdale, Pennsylvania 19446
| | - Matthew G Hartman
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Heather K Schofield
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Kevin P Stavrides
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Danielle E Talbot
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111 St. Hubert Catholic High School for Girls, Philadelphia, Pennsylvania 19136
| | - Devon J Riggs
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111 Science Scholars Program, Temple University, Philadelphia, Pennsylvania 19122
| | - Caroline Pritchard
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111 Souderton Area High School, Souderton, Pennsylvania 18964
| | - Alana M O'Reilly
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| |
Collapse
|
33
|
Vlachos S, Jangam S, Conder R, Chou M, Nystul T, Harden N. A Pak-regulated cell intercalation event leading to a novel radial cell polarity is involved in positioning of the follicle stem cell niche in the Drosophila ovary. Development 2015; 142:82-91. [PMID: 25516970 DOI: 10.1242/dev.111039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the germarium of the Drosophila ovary, germline cysts are encapsulated one at a time by a follicular epithelium derived from two follicle stem cells (FSCs). Ovaries in flies mutant for the serine/threonine kinase Pak exhibit a novel phenotype, in which two side-by-side cysts are encapsulated at a time, generating paired egg chambers. This striking phenotype originates in the pupal ovary, where the developing germarium is shaped by the basal stalk, a stack of cells formed by cell intercalation. The process of basal stalk formation is not well understood, and we provide evidence that the cell intercalation is driven by actomyosin contractility of DE-Cadherin-adhered cells, leading to a column of disk-shaped cells exhibiting a novel radial cell polarity. Cell intercalation fails in Pak mutant ovaries, leading to abnormally wide basal stalks and consequently wide germaria with side-by-side cysts. We present evidence that Pak mutant germaria have extra FSCs, and we propose that contact of a germline cyst with the basal stalk in the pupal ovary contributes to FSC niche formation. The wide basal stalk in Pak mutants enables the formation of extra FSC niches which are mispositioned and yet functional, indicating that the FSC niche can be established in diverse locations.
Collapse
Affiliation(s)
- Stephanie Vlachos
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Sharayu Jangam
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Ryan Conder
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Michael Chou
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Todd Nystul
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6 Departments of Anatomy and OB/GYN-RS, University of California, San Francisco, CA 94143, USA
| | - Nicholas Harden
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| |
Collapse
|
34
|
Maartens AP, Brown NH. Anchors and signals: the diverse roles of integrins in development. Curr Top Dev Biol 2015; 112:233-72. [PMID: 25733142 DOI: 10.1016/bs.ctdb.2014.11.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Integrins mediate cell adhesion by providing a link between the actin cytoskeleton and the extracellular matrix. As well as acting to anchor cells, integrin adhesions provide sensory input via mechanotransduction and synergism with signaling pathways, and provide the cell with the conditions necessary for differentiation in a permissive manner. In this review, we explore how integrins contribute to development, and what this tells us about how they work. From a signaling perspective, the influence of integrins on cell viability and fate is muted in a developmental context as compared to cell culture. Integrin phenotypes tend to arise from a failure of normally specified cells to create tissues properly, due to defective adhesion. The diversity of integrin functions in development shows how cell adhesion is continuously adjusted, both within and between animals, to fit developmental purpose.
Collapse
Affiliation(s)
- Aidan P Maartens
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
35
|
Wang X, Page-McCaw A. A matrix metalloproteinase mediates long-distance attenuation of stem cell proliferation. ACTA ACUST UNITED AC 2014; 206:923-36. [PMID: 25267296 PMCID: PMC4178971 DOI: 10.1083/jcb.201403084] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Long-range signaling by Wingless in the Drosophila ovary requires the glypican Dally-like and is antagonized by Dally-like cleavage by the extracellular metalloproteinase Mmp2. Ligand-based signaling can potentiate communication between neighboring cells and between cells separated by large distances. In the Drosophila melanogaster ovary, Wingless (Wg) promotes proliferation of follicle stem cells located ∼50 µm or five cell diameters away from the Wg source. How Wg traverses this distance is unclear. We find that this long-range signaling requires Division abnormally delayed (Dally)-like (Dlp), a glypican known to extend the range of Wg ligand in the wing disc by binding Wg. Dlp-mediated spreading of Wg to follicle stem cells is opposed by the extracellular protease Mmp2, which cleaved Dlp in cell culture, triggering its relocalization such that Dlp no longer contacted Wg protein. Mmp2-deficient ovaries displayed increased Wg distribution, activity, and stem cell proliferation. Mmp2 protein is expressed in the same cells that produce Wg; thus, niche cells produce both a long-range stem cell proliferation factor and a negative regulator of its spreading. This system could allow for spatial control of Wg signaling to targets at different distances from the source.
Collapse
Affiliation(s)
- Xiaoxi Wang
- Department of Cell and Developmental Biology, Program in Developmental Biology, and Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Department of Cell and Developmental Biology, Program in Developmental Biology, and Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology, Program in Developmental Biology, and Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Department of Cell and Developmental Biology, Program in Developmental Biology, and Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Department of Cell and Developmental Biology, Program in Developmental Biology, and Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
36
|
Pleiotropy of the Drosophila JAK pathway cytokine Unpaired 3 in development and aging. Dev Biol 2014; 395:218-31. [PMID: 25245869 DOI: 10.1016/j.ydbio.2014.09.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 09/10/2014] [Accepted: 09/12/2014] [Indexed: 12/22/2022]
Abstract
The Janus kinase (JAK) pathway is an essential, highly re-utilized developmental signaling cascade found in most metazoans. In vertebrates, the JAK intracellular cascade mediates signaling by dozens of cytokines and growth factors. In Drosophila, the Unpaired (Upd) family, encoded by three tandemly duplicated genes, is the only class of ligands associated with JAK stimulation. Unpaired has a central role in activation of JAK for most pathway functions, while Unpaired 2 regulates body size through insulin signaling. We show here that the third member of the family, unpaired 3 (upd3), overlaps upd in expression in some tissues and is essential for a subset of JAK-mediated developmental functions. First, consistent with the known requirements of JAK signaling in gametogenesis, we find that mutants of upd3 show an age-dependent impairment of fertility in both sexes. In oogenesis, graded JAK activity stimulated by Upd specifies the fates of the somatic follicle cells. As upd3 mutant females age, defects arise that can be attributed to perturbations of the terminal follicle cells, which require the highest levels of JAK activation. Therefore, in oogenesis, the activities of Upd and Upd3 both appear to quantitatively contribute to specification of those follicle cell fates. Furthermore, the sensitization of upd3 mutants to age-related decline in fertility can be used to investigate reproductive senescence. Second, loss of Upd3 during imaginal development results in defects of adult structures, including reduced eye size and abnormal wing and haltere posture. The outstretched wing and small eye phenotypes resemble classical alleles referred to as outstretched (os) mutations that have been previously ascribed to upd. However, we show that os alleles affect expression of both upd and upd3 and map to untranscribed regions, suggesting that they disrupt regulatory elements shared by both genes. Thus the upd region serves as a genetically tractable model for coordinate regulation of tandemly duplicated gene families that are commonly found in higher eukaryotes.
Collapse
|
37
|
Inhibiting stromal cell heparan sulfate synthesis improves stem cell mobilization and enables engraftment without cytotoxic conditioning. Blood 2014; 124:2937-47. [PMID: 25202142 DOI: 10.1182/blood-2014-08-593426] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The glycosyltransferase gene, Ext1, is essential for heparan sulfate production. Induced deletion of Ext1 selectively in Mx1-expressing bone marrow (BM) stromal cells, a known population of skeletal stem/progenitor cells, in adult mice resulted in marked changes in hematopoietic stem and progenitor cell (HSPC) localization. HSPC egressed from BM to spleen after Ext1 deletion. This was associated with altered signaling in the stromal cells and with reduced vascular cell adhesion molecule 1 production by them. Further, pharmacologic inhibition of heparan sulfate mobilized qualitatively more potent and quantitatively more HSPC from the BM than granulocyte colony-stimulating factor alone, including in a setting of granulocyte colony-stimulating factor resistance. The reduced presence of endogenous HSPC after Ext1 deletion was associated with engraftment of transfused HSPC without any toxic conditioning of the host. Therefore, inhibiting heparan sulfate production may provide a means for avoiding the toxicities of radiation or chemotherapy in HSPC transplantation for nonmalignant conditions.
Collapse
|
38
|
Hsu YC, Li L, Fuchs E. Transit-amplifying cells orchestrate stem cell activity and tissue regeneration. Cell 2014; 157:935-49. [PMID: 24813615 DOI: 10.1016/j.cell.2014.02.057] [Citation(s) in RCA: 280] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/16/2014] [Accepted: 02/24/2014] [Indexed: 12/17/2022]
Abstract
Transit-amplifying cells (TACs) are an early intermediate in tissue regeneration. Here, using hair follicles (HFs) as a paradigm, we show that emerging TACs constitute a signaling center that orchestrates tissue growth. Whereas primed stem cells (SCs) generate TACs, quiescent SCs only proliferate after TACs form and begin expressing Sonic Hedgehog (SHH). TAC generation is independent of autocrine SHH, but the TAC pool wanes if they can't produce SHH. We trace this paradox to two direct actions of SHH: promoting quiescent-SC proliferation and regulating dermal factors that stoke TAC expansion. Ingrained within quiescent SCs' special sensitivity to SHH signaling is their high expression of GAS1. Without sufficient input from quiescent SCs, replenishment of primed SCs for the next hair cycle is compromised, delaying regeneration and eventually leading to regeneration failure. Our findings unveil TACs as transient but indispensable integrators of SC niche components and reveal an intriguing interdependency of primed and quiescent SC populations on tissue regeneration.
Collapse
Affiliation(s)
- Ya-Chieh Hsu
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Lishi Li
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
39
|
Gheorghisan-Galateanu AA, Hinescu ME, Enciu AM. Ovarian adult stem cells: hope or pitfall? J Ovarian Res 2014; 7:71. [PMID: 25018783 PMCID: PMC4094411 DOI: 10.1186/1757-2215-7-71] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/29/2014] [Indexed: 12/22/2022] Open
Abstract
For many years, ovarian biology has been based on the dogma that oocytes reserve in female mammals included a finite number, established before or at birth and it is determined by the number and quality of primordial follicles developed during the neonatal period. The restricted supply of oocytes in adult female mammals has been disputed in recent years by supporters of postnatal neo-oogenesis. Recent experimental data showed that ovarian surface epithelium and cortical tissue from both mouse and human were proved to contain very low proportion of cells able to propagate themselves, but also to generate immature oocytes in vitro or in vivo, when transplanted into immunodeficient mice ovaries. By mentioning several landmarks of ovarian stem cell reserve and addressing the exciting perspective of translation into clinical practice as treatment for infertility pathologies, the purpose of this article is to review the knowledge about adult mammalian ovarian stem cells, a topic that, since the first approach quickly attracted the attention of both the scientific media and patients.
Collapse
Affiliation(s)
- Ancuta Augustina Gheorghisan-Galateanu
- Department of Cellular and Molecular Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania ; C.I.Parhon National Institute of Endocrinology, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| | - Mihail Eugen Hinescu
- Department of Cellular and Molecular Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania ; V.Babes National Institute of Pathology, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| | - Ana Maria Enciu
- Department of Cellular and Molecular Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania ; V.Babes National Institute of Pathology, 8 Eroii Sanitari Blvd., 050474 Bucharest, Romania
| |
Collapse
|
40
|
Huang J, Kalderon D. Coupling of Hedgehog and Hippo pathways promotes stem cell maintenance by stimulating proliferation. ACTA ACUST UNITED AC 2014; 205:325-38. [PMID: 24798736 PMCID: PMC4018789 DOI: 10.1083/jcb.201309141] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It is essential to define the mechanisms by which external signals regulate adult stem cell numbers, stem cell maintenance, and stem cell proliferation to guide regenerative stem cell therapies and to understand better how cancers originate in stem cells. In this paper, we show that Hedgehog (Hh) signaling in Drosophila melanogaster ovarian follicle stem cells (FSCs) induces the activity of Yorkie (Yki), the transcriptional coactivator of the Hippo pathway, by inducing yki transcription. Moreover, both Hh signaling and Yki positively regulate the rate of FSC proliferation, both are essential for FSC maintenance, and both promote increased FSC longevity and FSC duplication when in excess. We also found that responses to activated Yki depend on Cyclin E induction while responses to excess Hh signaling depend on Yki induction, and excess Yki can compensate for defective Hh signaling. These causal connections provide the most rigorous evidence to date that a niche signal can promote stem cell maintenance principally by stimulating stem cell proliferation.
Collapse
Affiliation(s)
- Jianhua Huang
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | | |
Collapse
|
41
|
Berns N, Woichansky I, Friedrichsen S, Kraft N, Riechmann V. A genome-scale in vivo RNAi analysis of epithelial development in Drosophila identifies new proliferation domains outside of the stem cell niche. J Cell Sci 2014; 127:2736-48. [PMID: 24762813 DOI: 10.1242/jcs.144519] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Drosophila oogenesis system provides an excellent model to study the development of epithelial tissues. Here, we report the first genome-scale in vivo RNA interference (RNAi) screen for genes controlling epithelial development. By directly analysing cell and tissue architecture we identified 1125 genes, which we assigned to seven different functions in epithelial formation and homeostasis. We validated the significance of our screen by generating mutants for Vps60, a component of the endosomal sorting complexes required for transport (ESCRT) machinery. This analysis provided new insights into spatiotemporal control of cell proliferation in the follicular epithelium. Previous studies have identified signals controlling divisions in the follicle stem cell niche. However, 99% of cell divisions occur outside of the niche and it is unclear how these divisions are controlled. Our data distinguish two new domains outside of the stem cell niche where there are differing controls on proliferation. One domain abuts the niche and is characterised by ESCRT, Notch and JAK/STAT-mediated control of proliferation. Adjacent to this domain, another domain is defined by loss of the impact of ESCRT on cell division. Thus, during development epithelial cells pass through a variety of microenvironments that exert different modes of proliferation control. The switch between these modes might reflect a decrease in the 'stemness' of epithelial cells over time.
Collapse
Affiliation(s)
- Nicola Berns
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Innokenty Woichansky
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Steffen Friedrichsen
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Nadine Kraft
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Veit Riechmann
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology and German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
42
|
Rosales-Nieves AE, González-Reyes A. Genetics and mechanisms of ovarian cancer: parallels between Drosophila and humans. Semin Cell Dev Biol 2014; 28:104-9. [PMID: 24704277 DOI: 10.1016/j.semcdb.2014.03.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/26/2014] [Indexed: 02/05/2023]
Abstract
Considering the degree of detail available at the genetic and cellular levels, the Drosophila ovary stands out as a powerful system to identify new players in the regulation of key aspects of cancer progression. In this review, we will comment on how the use of the Drosophila ovary has helped to elucidate some of the molecular bases of ovarian malignancies and to identify and characterize critical tumour suppressor genes and oncogenes with an impact in human pathologies.
Collapse
Affiliation(s)
- Alicia E Rosales-Nieves
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC/JA, Carretera de Utrera km 1, 41013 Sevilla, Spain
| | - Acaimo González-Reyes
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC/JA, Carretera de Utrera km 1, 41013 Sevilla, Spain.
| |
Collapse
|
43
|
Rojas-Ríos P, González-Reyes A. Concise Review: The Plasticity of Stem Cell Niches: A General Property Behind Tissue Homeostasis and Repair. Stem Cells 2014; 32:852-9. [DOI: 10.1002/stem.1621] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/30/2013] [Accepted: 11/09/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Patricia Rojas-Ríos
- Centro Andaluz de Biología del Desarrollo; CSIC/Universidad Pablo de Olavide/JA; Carretera de Utrera km 1 Sevilla Spain
| | - Acaimo González-Reyes
- Centro Andaluz de Biología del Desarrollo; CSIC/Universidad Pablo de Olavide/JA; Carretera de Utrera km 1 Sevilla Spain
| |
Collapse
|
44
|
Sahai-Hernandez P, Nystul TG. A dynamic population of stromal cells contributes to the follicle stem cell niche in the Drosophila ovary. Development 2013; 140:4490-8. [PMID: 24131631 DOI: 10.1242/dev.098558] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Epithelial stem cells are maintained within niches that promote self-renewal by providing signals that specify the stem cell fate. In the Drosophila ovary, epithelial follicle stem cells (FSCs) reside in niches at the anterior tip of the tissue and support continuous growth of the ovarian follicle epithelium. Here, we demonstrate that a neighboring dynamic population of stromal cells, called escort cells, are FSC niche cells. We show that escort cells produce both Wingless and Hedgehog ligands for the FSC lineage, and that Wingless signaling is specific for the FSC niche whereas Hedgehog signaling is active in both FSCs and daughter cells. In addition, we show that multiple escort cells simultaneously encapsulate germ cell cysts and contact FSCs. Thus, FSCs are maintained in a dynamic niche by a non-dedicated population of niche cells.
Collapse
Affiliation(s)
- Pankaj Sahai-Hernandez
- Center for Reproductive Sciences, Departments of Anatomy and OB/GYN-RS, University of California, San Francisco, CA 94143-0452, USA
| | | |
Collapse
|
45
|
Raynaud CM, Butler JM, Halabi NM, Ahmad FS, Ahmed B, Rafii S, Rafii A. Endothelial cells provide a niche for placental hematopoietic stem/progenitor cell expansion through broad transcriptomic modification. Stem Cell Res 2013; 11:1074-90. [PMID: 23978474 DOI: 10.1016/j.scr.2013.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 07/17/2013] [Accepted: 07/31/2013] [Indexed: 11/26/2022] Open
Abstract
Umbilical cord blood (UCB) is an attractive source of hematopoietic stem cells (HSCs). However, the number of HSCs in UCB is limited, and attempts to amplify them in vitro remain inefficient. Several publications have documented amplification of hematopoietic stem/progenitor cells (HSPCs) on endothelial or mesenchymal cells, but the lack of homogeneity in culture conditions and HSC definition impairs direct comparison of these results. We investigated the ability of different feeder layers, mesenchymal progenitors (MPs) and endothelial cells (ECs), to amplify hematopoietic stem/progenitor cells. Placental derived HSPCs (defined as Lin(-)CD45(-/dim)CD34(+)CD38(-)CD90(+)) were maintained on confluent feeder layers and the number of cells and their marker expression were monitored over 21 days. Although both types of feeder layers supported hematopoietic expansion, only endothelial cells triggered amplification of Lin(-)CD45(-/dim)CD34(+)CD38(-)CD90(+) cells, which peaked at 14 days. The amplified cells differentiated into all cell lineages, as attested by in vitro colony-forming assays, and were capable of engraftment and multi-lineage differentiation in sub-lethally irradiated mice. Mesenchymal progenitors promoted amplification of CD38(+) cells, previously defined as precursors with more limited differentiation potential. A competitive assay demonstrated that hematopoietic stem/progenitor cells had a preference for interacting with endothelial cells in vitro. Cytokine and transcriptomic analysis of both feeder cell types identified differences in gene expression that correlated with propensity of ECs and MPs to support hematopoietic cell amplification and differentiation respectively. Finally, we used RNA sequencing of endothelial cells and HSPCs to uncover relevant networks illustrating the complex interaction between endothelial cells and HSPCs leading to stem/progenitor cell expansion.
Collapse
Affiliation(s)
- Christophe M Raynaud
- Qatar Cardiovascular Research Center, Qatar Foundation, Qatar Science and Technology Park, Doha, Qatar
| | | | | | | | | | | | | |
Collapse
|
46
|
Bausek N. JAK-STAT signaling in stem cells and their niches in Drosophila. JAKSTAT 2013; 2:e25686. [PMID: 24069566 PMCID: PMC3772118 DOI: 10.4161/jkst.25686] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/09/2013] [Accepted: 07/09/2013] [Indexed: 12/30/2022] Open
Abstract
JAK-STAT signaling is a highly conserved regulator of stem cells and their niches. Aberrant activation in hematopoietic stem cells is the underlying cause of a majority of myeloproliferative diseases. This review will focus on the roles of JAK-STAT activity in three different adult stem cell systems in Drosophila. Tightly controlled levels of JAK-STAT signaling are required for stem cell maintenance and self-renewal, as hyperactivation of the pathway is associated with stem cell overproliferation. JAK-STAT activity is further essential for anchoring the stem cells in their respective niches by regulating different adhesion molecules.
Collapse
Affiliation(s)
- Nina Bausek
- MRC Centre for Development and Biomedical Genetics and The Department of Biomedical Science; The University of Sheffield; Sheffield, UK
| |
Collapse
|
47
|
Hedgehog in the Drosophila testis niche: what does it do there? Protein Cell 2013; 4:650-5. [PMID: 23807635 DOI: 10.1007/s13238-013-3040-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 06/02/2013] [Indexed: 12/30/2022] Open
Abstract
Stem cell niche is a specialized microenvironment crucial to self-renewal. The testis in Drosophila contains two different types of stem cells, the germline stem cells and the somatic cyst stem cells that are sustained by their respective niche signals, thus is a good system for studying the interaction between the stem cells and their hosting niche. The JAK-STAT and BMP pathways are known to play critical roles in the self-renewal of different kinds of stem cells, but the roles of several other pathways have emerged recently in a complex signaling network in the testis niche. Reports of independent observations from three research groups have uncovered an important role of Hedgehog (Hh) in the Drosophila testis niche. In this review, we summarize these recent findings and discuss the interplay between the Hh signaling mechanisms and those of the JAK-STAT and BMP pathways. We also discuss directions for further investigation.
Collapse
|
48
|
Hartman TR, Strochlic TI, Ji Y, Zinshteyn D, O'Reilly AM. Diet controls Drosophila follicle stem cell proliferation via Hedgehog sequestration and release. ACTA ACUST UNITED AC 2013; 201:741-57. [PMID: 23690177 PMCID: PMC3664720 DOI: 10.1083/jcb.201212094] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dietary cholesterol levels control follicle stem cell proliferation in the Drosophila ovary via regulation of Hedgehog protein localization. A healthy diet improves adult stem cell function and delays diseases such as cancer, heart disease, and neurodegeneration. Defining molecular mechanisms by which nutrients dictate stem cell behavior is a key step toward understanding the role of diet in tissue homeostasis. In this paper, we elucidate the mechanism by which dietary cholesterol controls epithelial follicle stem cell (FSC) proliferation in the fly ovary. In nutrient-restricted flies, the transmembrane protein Boi sequesters Hedgehog (Hh) ligand at the surface of Hh-producing cells within the ovary, limiting FSC proliferation. Upon feeding, dietary cholesterol stimulates S6 kinase–mediated phosphorylation of the Boi cytoplasmic domain, triggering Hh release and FSC proliferation. This mechanism enables a rapid, tissue-specific response to nutritional changes, tailoring stem cell divisions and egg production to environmental conditions sufficient for progeny survival. If conserved in other systems, this mechanism will likely have important implications for studies on molecular control of stem cell function, in which the benefits of low calorie and low cholesterol diets are beginning to emerge.
Collapse
Affiliation(s)
- Tiffiney R Hartman
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | |
Collapse
|