1
|
Chen R, Grill S, Lin B, Saiduddin M, Lehmann R. Origin and establishment of the germline in Drosophila melanogaster. Genetics 2025; 229:iyae217. [PMID: 40180587 PMCID: PMC12005264 DOI: 10.1093/genetics/iyae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/09/2024] [Indexed: 04/05/2025] Open
Abstract
The continuity of a species depends on germ cells. Germ cells are different from all the other cell types of the body (somatic cells) as they are solely destined to develop into gametes (sperm or egg) to create the next generation. In this review, we will touch on 4 areas of embryonic germ cell development in Drosophila melanogaster: the assembly and function of germplasm, which houses the determinants for germ cell specification and fate and the mitochondria of the next generation; the process of pole cell formation, which will give rise to primordial germ cells (PGCs); the specification of pole cells toward the PGC fate; and finally, the migration of PGCs to the somatic gonadal precursors, where they, together with somatic gonadal precursors, form the embryonic testis and ovary.
Collapse
Affiliation(s)
- Ruoyu Chen
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Vilcek Institute of Graduate Studies, Department of Cell Biology, NYU School of Medicine, New York University, New York, NY 10016, USA
| | - Sherilyn Grill
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Benjamin Lin
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mariyah Saiduddin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Vilcek Institute of Graduate Studies, Department of Cell Biology, NYU School of Medicine, New York University, New York, NY 10016, USA
| | - Ruth Lehmann
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
2
|
Barton LJ, Roa-de la Cruz L, Lehmann R, Lin B. The journey of a generation: advances and promises in the study of primordial germ cell migration. Development 2024; 151:dev201102. [PMID: 38607588 PMCID: PMC11165723 DOI: 10.1242/dev.201102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
The germline provides the genetic and non-genetic information that passes from one generation to the next. Given this important role in species propagation, egg and sperm precursors, called primordial germ cells (PGCs), are one of the first cell types specified during embryogenesis. In fact, PGCs form well before the bipotential somatic gonad is specified. This common feature of germline development necessitates that PGCs migrate through many tissues to reach the somatic gonad. During their journey, PGCs must respond to select environmental cues while ignoring others in a dynamically developing embryo. The complex multi-tissue, combinatorial nature of PGC migration is an excellent model for understanding how cells navigate complex environments in vivo. Here, we discuss recent findings on the migratory path, the somatic cells that shepherd PGCs, the guidance cues somatic cells provide, and the PGC response to these cues to reach the gonad and establish the germline pool for future generations. We end by discussing the fate of wayward PGCs that fail to reach the gonad in diverse species. Collectively, this field is poised to yield important insights into emerging reproductive technologies.
Collapse
Affiliation(s)
- Lacy J. Barton
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Lorena Roa-de la Cruz
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Ruth Lehmann
- Whitehead Institute and Department of Biology, MIT, 455 Main Street, Cambridge, MA 02142, USA
| | - Benjamin Lin
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| |
Collapse
|
3
|
Deshpande G, Ng C, Jourjine N, Chiew JW, Dasilva J, Schedl P. Hedgehog signaling guides migration of primordial germ cells to the Drosophila somatic gonad. Genetics 2023; 225:iyad165. [PMID: 37708366 PMCID: PMC10627259 DOI: 10.1093/genetics/iyad165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023] Open
Abstract
In addition to inducing nonautonomous specification of cell fate in both Drosophila and vertebrates, the Hedgehog pathway guides cell migration in a variety of different tissues. Although its role in axon guidance in the vertebrate nervous system is widely recognized, its role in guiding the migratory path of primordial germ cells (PGCs) from the outside surface of the Drosophila embryo through the midgut and mesoderm to the SGPs (somatic gonadal precursors) has been controversial. Here we present new experiments demonstrating (1) that Hh produced by mesodermal cells guides PGC migration, (2) that HMG CoenzymeA reductase (Hmgcr) potentiates guidance signals emanating from the SGPs, functioning upstream of hh and of 2 Hh pathway genes important for Hh-containing cytonemes, and (3) that factors required in Hh receiving cells in other contexts function in PGCs to help direct migration toward the SGPs. We also compare the data reported by 4 different laboratories that have studied the role of the Hh pathway in guiding PGC migration.
Collapse
Affiliation(s)
- Girish Deshpande
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Chris Ng
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Nicholas Jourjine
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Joy Wan Chiew
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Juliana Dasilva
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Paul Schedl
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
4
|
Sustar AE, Strand LG, Zimmerman SG, Berg CA. Imaginal disk growth factors are Drosophila chitinase-like proteins with roles in morphogenesis and CO2 response. Genetics 2023; 223:iyac185. [PMID: 36576887 PMCID: PMC9910413 DOI: 10.1093/genetics/iyac185] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 07/18/2022] [Accepted: 11/16/2022] [Indexed: 12/29/2022] Open
Abstract
Chitinase-like proteins (CLPs) are members of the family 18 glycosyl hydrolases, which include chitinases and the enzymatically inactive CLPs. A mutation in the enzyme's catalytic site, conserved in vertebrates and invertebrates, allowed CLPs to evolve independently with functions that do not require chitinase activity. CLPs normally function during inflammatory responses, wound healing, and host defense, but when they persist at excessive levels at sites of chronic inflammation and in tissue-remodeling disorders, they correlate positively with disease progression and poor prognosis. Little is known, however, about their physiological function. Drosophila melanogaster has 6 CLPs, termed Imaginal disk growth factors (Idgfs), encoded by Idgf1, Idgf2, Idgf3, Idgf4, Idgf5, and Idgf6. In this study, we developed tools to facilitate characterization of the physiological roles of the Idgfs by deleting each of the Idgf genes using the CRISPR/Cas9 system and assessing loss-of-function phenotypes. Using null lines, we showed that loss of function for all 6 Idgf proteins significantly lowers viability and fertility. We also showed that Idgfs play roles in epithelial morphogenesis, maintaining proper epithelial architecture and cell shape, regulating E-cadherin and cortical actin, and remarkably, protecting these tissues against CO2 exposure. Defining the normal molecular mechanisms of CLPs is a key to understanding how deviations tip the balance from a physiological to a pathological state.
Collapse
Affiliation(s)
- Anne E Sustar
- Department of Genome Sciences, University of Washington, Foege Bldg. S-250, 3720 15th Ave NE, Seattle, WA 98195-5065, USA
| | - Liesl G Strand
- Department of Genome Sciences, University of Washington, Foege Bldg. S-250, 3720 15th Ave NE, Seattle, WA 98195-5065, USA
| | - Sandra G Zimmerman
- Department of Genome Sciences, University of Washington, Foege Bldg. S-250, 3720 15th Ave NE, Seattle, WA 98195-5065, USA
| | - Celeste A Berg
- Department of Genome Sciences, University of Washington, Foege Bldg. S-250, 3720 15th Ave NE, Seattle, WA 98195-5065, USA
| |
Collapse
|
5
|
Transcriptional Inhibition of Sp-IAG by Crustacean Female Sex Hormone in the Mud Crab, Scylla paramamosain. Int J Mol Sci 2020; 21:ijms21155300. [PMID: 32722594 PMCID: PMC7432471 DOI: 10.3390/ijms21155300] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 01/09/2023] Open
Abstract
In crustaceans, the regulation of sex differentiation is mediated by insulin-like androgenic hormone (IAG) and crustacean female sex hormone (CFSH). CFSH is reported to inhibit IAG gene (Sp-IAG) expression in the mud crab Scylla paramamosain, but the regulatory mechanism is not well understood. A 2674 bp 5′ flanking Sp-IAG contains many potential transcription factor binding sites. In this study, analysis of serially deleted 5′ flanking Sp-IAG and site-directed mutation (SDM) of transcription factor binding sites of the same gene showed that the promoter activity of reporter vectors with Sox-5-binding site, signal transducers and activators of transcription (STAT)-binding site and activator protein 1 (AP-1)-binding site were significantly higher than that of vectors without these regions, suggesting that they were involved in transcriptional regulation of Sp-IAG expression. The expression analysis of these transcription factor showed that there was no difference in the level of mRNA in Sox-5 and AP-1 in androgenic gland treated with recombinant CFSH, but expression of Sp-STAT was significantly reduced, suggesting that CFSH regulates the expression of Sp-STAT, inhibiting its function to regulate Sp-IAG. Further experiment revealed that RNAi mediated Sp-STAT gene knockdown reduced the expression of Sp-IAG. These results suggested that Sp-CFSH regulates Sp-IAG by inhibiting STAT. This is a pioneering finding on the transcriptional mechanism of IAG gene in crustaceans.
Collapse
|
6
|
Tan K, Song HW, Wilkinson MF. Single-cell RNAseq analysis of testicular germ and somatic cell development during the perinatal period. Development 2020; 147:dev.183251. [PMID: 31964773 DOI: 10.1242/dev.183251] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/03/2020] [Indexed: 12/22/2022]
Abstract
Pro-spermatogonia (SG) serve as the gateway to spermatogenesis. Using single-cell RNA sequencing (RNAseq), we studied the development of ProSG, their SG descendants and testicular somatic cells during the perinatal period in mice. We identified both gene and protein markers for three temporally distinct ProSG cell subsets, including a migratory cell population with a transcriptome distinct from the previously defined T1- and T2-ProSG stages. This intermediate (I)-ProSG subset translocates from the center of seminiferous tubules to the spermatogonial stem cell (SSC) 'niche' in its periphery soon after birth. We identified three undifferentiated SG subsets at postnatal day 7, each of which expresses distinct genes, including transcription factor and signaling genes. Two of these subsets have the characteristics of newly emergent SSCs. We also molecularly defined the development of Sertoli, Leydig and peritubular myoid cells during the perinatal period, allowing us to identify candidate signaling pathways acting between somatic and germ cells in a stage-specific manner during the perinatal period. Our study provides a rich resource for those investigating testicular germ and somatic cell developmental during the perinatal period.
Collapse
Affiliation(s)
- Kun Tan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Hye-Won Song
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Miles F Wilkinson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA .,Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Tettweiler G, Blaquiere JA, Wray NB, Verheyen EM. Hipk is required for JAK/STAT activity during development and tumorigenesis. PLoS One 2019; 14:e0226856. [PMID: 31891940 PMCID: PMC6938406 DOI: 10.1371/journal.pone.0226856] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/05/2019] [Indexed: 12/24/2022] Open
Abstract
Drosophila has been instrumental as a model system in studying signal transduction and revealing molecular functions in development and human diseases. A point mutation in the Drosophila Janus kinase JAK (called hop) causes constitutive activation of the JAK/STAT pathway. We provide robust genetic evidence that the Homeodomain interacting protein kinase (Hipk) is required for endogenous JAK/STAT activity. Overexpression of Hipk can phenocopy the effects of overactive JAK/STAT mutations and lead to melanized tumors, and loss of Hipk can suppress the effects of hyperactive JAK/STAT. Further, the loss of the pathway effector Stat92E can suppress Hipk induced overgrowth. Interaction studies show that Hipk can physically interact with Stat92E and regulate Stat92E subcellular localization. Together our results show that Hipk is a novel factor required for effective JAK/STAT signaling.
Collapse
Affiliation(s)
- Gritta Tettweiler
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, B.C Canada
| | - Jessica A. Blaquiere
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, B.C Canada
| | - Nathan B. Wray
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, B.C Canada
| | - Esther M. Verheyen
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, B.C Canada
- * E-mail:
| |
Collapse
|
8
|
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: 131] [Impact Index Per Article: 21.8] [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
|
9
|
Trivedi S, Starz-Gaiano M. Drosophila Jak/STAT Signaling: Regulation and Relevance in Human Cancer and Metastasis. Int J Mol Sci 2018; 19:ijms19124056. [PMID: 30558204 PMCID: PMC6320922 DOI: 10.3390/ijms19124056] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/08/2018] [Accepted: 12/11/2018] [Indexed: 12/26/2022] Open
Abstract
Over the past three-decades, Janus kinase (Jak) and signal transducer and activator of transcription (STAT) signaling has emerged as a paradigm to understand the involvement of signal transduction in development and disease pathology. At the molecular level, cytokines and interleukins steer Jak/STAT signaling to transcriptional regulation of target genes, which are involved in cell differentiation, migration, and proliferation. Jak/STAT signaling is involved in various types of blood cell disorders and cancers in humans, and its activation is associated with carcinomas that are more invasive or likely to become metastatic. Despite immense information regarding Jak/STAT regulation, the signaling network has numerous missing links, which is slowing the progress towards developing drug therapies. In mammals, many components act in this cascade, with substantial cross-talk with other signaling pathways. In Drosophila, there are fewer pathway components, which has enabled significant discoveries regarding well-conserved regulatory mechanisms. Work across species illustrates the relevance of these regulators in humans. In this review, we showcase fundamental Jak/STAT regulation mechanisms in blood cells, stem cells, and cell motility. We examine the functional relevance of key conserved regulators from Drosophila to human cancer stem cells and metastasis. Finally, we spotlight less characterized regulators of Drosophila Jak/STAT signaling, which stand as promising candidates to be investigated in cancer biology. These comparisons illustrate the value of using Drosophila as a model for uncovering the roles of Jak/STAT signaling and the molecular means by which the pathway is controlled.
Collapse
Affiliation(s)
- Sunny Trivedi
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Michelle Starz-Gaiano
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
10
|
JAK-STAT signaling regulation of chicken embryonic stem cell differentiation into male germ cells. In Vitro Cell Dev Biol Anim 2017; 53:728-743. [PMID: 28597334 DOI: 10.1007/s11626-017-0167-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/26/2017] [Indexed: 12/14/2022]
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling is crucial in chicken germ stem cell differentiation, but its role in the regulation of germ cell differentiation is unknown. To address this, cucurbitacin I or interleukin 6 was used to inhibit or activate JAK-STAT signaling during embryonic stem cells (ESCs) differentiation. The expression of downstream JAK-STAT signaling molecules was assessed by Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR). PAS, and immunohistochemical staining of frozen sections was used to determine the appearance of primordial germ cells (PGCs) and, later, spermatogonial stem cells (SSCs) during gonadal development. Inhibition of the JAK-STAT signaling resulted in decreased expression of JAK2 and STAT3 as well as of PGCs markers; moreover, the proportion of CVH and C-KIT positive cells as well as the yield of PGCs were remarkably decreased, and the gonad was smaller than that of control samples. Conversely, activation of JAK-STAT resulted in increased expression of JAK2 and STAT3 as well as that of PGC marker CVH. In addition, the proportion of CVH and C-KIT-positive cells as well as the PGC yield was increased, and the gonad was significantly larger than that from control samples. Collectively, our results suggested that JAK-STAT effectively promoted the formation of PGCs in the genital ridge during early embryogenesis in vivo and played a positive role in the regulation of ESC to SSC differentiation in vitro, with JAK2 and STAT3 functioning as pivotal factors for intracellular signal transduction.
Collapse
|
11
|
Mills SC, Goh PH, Kudatsih J, Ncube S, Gurung R, Maxwell W, Mueller A. Cell migration towards CXCL12 in leukemic cells compared to breast cancer cells. Cell Signal 2016; 28:316-24. [DOI: 10.1016/j.cellsig.2016.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 01/19/2016] [Accepted: 01/19/2016] [Indexed: 12/23/2022]
|
12
|
Abstract
Leukemia inhibitory factor (LIF) is a member of the interleukin-6 (IL-6) cytokine family. All members of this family activate signal transducer and activator of transcription 3 (STAT3), a transcription factor that influences stem and progenitor cell identity, proliferation and cytoprotection. The role of LIF in development was first identified when LIF was demonstrated to support the propagation of mouse embryonic stem cells. Subsequent studies of mice deficient for components of the LIF pathway have revealed important roles for LIF signaling during development and homeostasis. Here and in the accompanying poster, we provide a broad overview of JAK-STAT signaling during development, with a specific focus on LIF-mediated JAK-STAT3 activation.
Collapse
Affiliation(s)
- Kento Onishi
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G9
| | - Peter W Zandstra
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G9 Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada M5S 3E5 The Donnelly Centre, University of Toronto, 160 College St., Toronto, Ontario, Canada M5S 3E1 McEwen Centre for Regenerative Medicine, University Health Network, 101 College St., Toronto, Ontario, Canada M5G 1L7
| |
Collapse
|
13
|
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
|
14
|
Chen Q, Giedt M, Tang L, Harrison DA. Tools and methods for studying the Drosophila JAK/STAT pathway. Methods 2014; 68:160-72. [DOI: 10.1016/j.ymeth.2014.03.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 03/17/2014] [Accepted: 03/19/2014] [Indexed: 12/29/2022] Open
|
15
|
Demarco RS, Eikenes ÅH, Haglund K, Jones DL. Investigating spermatogenesis in Drosophila melanogaster. Methods 2014; 68:218-27. [PMID: 24798812 DOI: 10.1016/j.ymeth.2014.04.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/24/2014] [Accepted: 04/25/2014] [Indexed: 01/05/2023] Open
Abstract
The process of spermatogenesis in Drosophila melanogaster provides a powerful model system to probe a variety of developmental and cell biological questions, such as the characterization of mechanisms that regulate stem cell behavior, cytokinesis, meiosis, and mitochondrial dynamics. Classical genetic approaches, together with binary expression systems, FRT-mediated recombination, and novel imaging systems to capture single cell behavior, are rapidly expanding our knowledge of the molecular mechanisms regulating all aspects of spermatogenesis. This methods chapter provides a detailed description of the system, a review of key questions that have been addressed or remain unanswered thus far, and an introduction to tools and techniques available to probe each stage of spermatogenesis.
Collapse
Affiliation(s)
- Rafael S Demarco
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Åsmund H Eikenes
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, 0379 Montebello, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, Oslo University Hospital, 0379 Montebello, Norway
| | - Kaisa Haglund
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, 0379 Montebello, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, Oslo University Hospital, 0379 Montebello, Norway
| | - D Leanne Jones
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
16
|
Mukherjee A, Neher RA, Renault AD. Quantifying the range of a lipid phosphate signal in vivo. J Cell Sci 2013; 126:5453-64. [PMID: 24006260 DOI: 10.1242/jcs.136176] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Quantitative information about the range of influence of extracellular signaling molecules is critical for understanding their effects, but is difficult to determine in the complex and dynamic three-dimensional environment of a living embryo. Drosophila germ cells migrate during embryogenesis and use spatial information provided by expression of lipid phosphate phosphatases called Wunens to reach the somatic gonad. However, whether guidance requires cell contact or involves a diffusible signal is not known. We ectopically expressed Wunens in various segmentally repeated ectodermal and parasegmental patterns in embryos otherwise null for Wunens and used germ cell behavior to show that the signal is diffusible and to define its range. We correlated this back to the wild-type scenario and found that the germ cell migratory path can be primarily accounted for by Wunen expression. This approach provides the first quantitative information of the effective range of a lipid phosphate in vivo and has implications for the migration of other cell types that respond to lipid phosphates.
Collapse
Affiliation(s)
- Amrita Mukherjee
- Max Planck Institute for Developmental Biology, Spemannstrasse 35, 72076 Tübingen, Germany
| | | | | |
Collapse
|
17
|
Hombría JCG, Sotillos S. JAK-STAT pathway in Drosophila morphogenesis: From organ selector to cell behavior regulator. JAKSTAT 2013; 2:e26089. [PMID: 24069568 PMCID: PMC3772120 DOI: 10.4161/jkst.26089] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 11/19/2022] Open
Abstract
One of the main contributions of Drosophila to the JAK-STAT field is the study of morphogenesis. JAK-STAT signaling controls the formation of many different structures through surprisingly different morphogenetic behaviors that include induction of cell rearrangements, invagination, folding of tissues, modulation of cell shape, and migration. This variability may be explained by the many transcription factors and signaling molecules STAT regulates at early stages of development. But is STAT just acting as an upstream inducer of morphogenesis or does it have a more direct role in controlling cell behaviors? Here we review what is known about how the canonical phosphorylation of STAT contributes to shaping the embryonic and imaginal structures.
Collapse
|
18
|
Quint P, Ruan M, Pederson L, Kassem M, Westendorf JJ, Khosla S, Oursler MJ. Sphingosine 1-phosphate (S1P) receptors 1 and 2 coordinately induce mesenchymal cell migration through S1P activation of complementary kinase pathways. J Biol Chem 2013; 288:5398-406. [PMID: 23300082 DOI: 10.1074/jbc.m112.413583] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Normal bone turnover requires tight coupling of bone resorption and bone formation to preserve bone quantity and structure. With aging and during several pathological conditions, this coupling breaks down, leading to either net bone loss or excess bone formation. To preserve or restore normal bone metabolism, it is crucial to determine the mechanisms by which osteoclasts and osteoblast precursors interact and contribute to coupling. We showed that osteoclasts produce the chemokine sphingosine 1-phosphate (S1P), which stimulates osteoblast migration. Thus, osteoclast-derived S1P may recruit osteoblasts to sites of bone resorption as an initial step in replacing lost bone. In this study we investigated the mechanisms by which S1P stimulates mesenchymal (skeletal) cell chemotaxis. S1P treatment of mesenchymal (skeletal) cells activated RhoA GTPase, but this small G protein did not contribute to migration. Rather, two S1P receptors, S1PR1 and S1PR2, coordinately promoted migration through activation of the JAK/STAT3 and FAK/PI3K/AKT signaling pathways, respectively. These data demonstrate that the chemokine S1P couples bone formation to bone resorption through activation of kinase signaling pathways.
Collapse
Affiliation(s)
- Patrick Quint
- Endocrine Research Unit and Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Group choreography: mechanisms orchestrating the collective movement of border cells. Nat Rev Mol Cell Biol 2012; 13:631-45. [PMID: 23000794 DOI: 10.1038/nrm3433] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cell movements are essential for animal development and homeostasis but also contribute to disease. Moving cells typically extend protrusions towards a chemoattractant, adhere to the substrate, contract and detach at the rear. It is less clear how cells that migrate in interconnected groups in vivo coordinate their behaviour and navigate through natural environments. The border cells of the Drosophila melanogaster ovary have emerged as an excellent model for the study of collective cell movement, aided by innovative genetic, live imaging, and photomanipulation techniques. Here we provide an overview of the molecular choreography of border cells and its more general implications.
Collapse
|
20
|
Khabbazi S, Jacques R, Moyano Cardaba C, Mueller A. Janus kinase 2 and signal transducer and activator of transcription 3 activation is not essential for CCL3-, CCL5- or CCL8-induced chemotaxis. Cell Biochem Funct 2012; 31:312-8. [DOI: 10.1002/cbf.2901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 08/28/2012] [Accepted: 08/30/2012] [Indexed: 01/08/2023]
Affiliation(s)
- S. Khabbazi
- School of Pharmacy; University of East Anglia; Norwich; UK
| | - R.O. Jacques
- School of Pharmacy; University of East Anglia; Norwich; UK
| | | | - A. Mueller
- School of Pharmacy; University of East Anglia; Norwich; UK
| |
Collapse
|
21
|
Colodner KJ, Feany MB. Glial fibrillary tangles and JAK/STAT-mediated glial and neuronal cell death in a Drosophila model of glial tauopathy. J Neurosci 2010; 30:16102-13. [PMID: 21123557 PMCID: PMC3073608 DOI: 10.1523/jneurosci.2491-10.2010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2010] [Revised: 09/15/2010] [Accepted: 09/17/2010] [Indexed: 11/21/2022] Open
Abstract
A subset of neurodegenerative tauopathies is characterized by abundant filamentous inclusions of hyperphosphorylated tau in both neurons and glia. Although the contribution of neuronal tau to behavioral changes and neuronal loss in neurodegenerative diseases has been studied extensively, the functional consequences of tau deposition in glial cells have been less well characterized. To investigate the role of abnormal tau accumulation and aggregation in glial cells, we created a Drosophila model of glial tauopathy by expressing human wild-type tau in adult fly glial cells. Glial expression of tau resulted in robust aggregation of phosphorylated tau into fibrillary inclusions similar to human glial tangles. Tangle formation was accompanied by shortened lifespan and age-dependent apoptotic cell death of both glia and neurons. Genetic manipulation of Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling modified toxicity of glial tau. We also identified a synergistic interaction of combined tau expression in neurons and glial cells. In summary, we present a genetically tractable model of glial fibrillary tau tangle formation and identify JAK/STAT signaling as mediating the death of both glia and neurons in this model.
Collapse
Affiliation(s)
- Kenneth J. Colodner
- Program in Neuroscience, Harvard Medical School, Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Mel B. Feany
- Program in Neuroscience, Harvard Medical School, Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115
| |
Collapse
|
22
|
Bertet C, Rauzi M, Lecuit T. Repression of Wasp by JAK/STAT signalling inhibits medial actomyosin network assembly and apical cell constriction in intercalating epithelial cells. Development 2010; 136:4199-212. [PMID: 19934015 DOI: 10.1242/dev.040402] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Tissue morphogenesis requires stereotyped cell shape changes, such as apical cell constriction in the mesoderm and cell intercalation in the ventrolateral ectoderm of Drosophila. Both processes require force generation by an actomyosin network. The subcellular localization of Myosin-II (Myo-II) dictates these different morphogenetic processes. In the intercalating ectoderm Myo-II is mostly cortical, but in the mesoderm Myo-II is concentrated in a medial meshwork. We report that apical constriction is repressed by JAK/STAT signalling in the lateral ectoderm independently of Twist. Inactivation of the JAK/STAT pathway causes germband extension defects because of apical constriction ventrolaterally. This is associated with ectopic recruitment of Myo-II in a medial web, which causes apical cell constriction as shown by laser nanosurgery. Reducing Myo-II levels rescues the JAK/STAT mutant phenotype, whereas overexpression of the Myo-II heavy chain (also known as Zipper), or constitutive activation of its regulatory light chain, does not cause medial accumulation of Myo-II nor apical constriction. Thus, JAK/STAT controls Myo-II localization by additional mechanisms. We show that regulation of actin polymerization by Wasp, but not by Dia, is important in this process. Constitutive activation of Wasp, a branched actin regulator, causes apical cell constriction and promotes medial 'web' formation. Wasp is inactivated at the cell cortex in the germband by JAK/STAT signalling. Lastly, wasp mutants rescue the normal cortical enrichment of Myo-II and inhibit apical constriction in JAK/STAT mutants, indicating that Wasp is an effector of JAK/STAT signalling in the germband. We discuss possible models for the role of Wasp activity in the regulation of Myo-II distribution.
Collapse
Affiliation(s)
- Claire Bertet
- IBDML, UMR6216 CNRS-Université de la Méditerranée, Campus de Luminy, case 907, 13288 Marseille Cedex 09, France
| | | | | |
Collapse
|
23
|
Richardson BE, Lehmann R. Mechanisms guiding primordial germ cell migration: strategies from different organisms. Nat Rev Mol Cell Biol 2010; 11:37-49. [PMID: 20027186 PMCID: PMC4521894 DOI: 10.1038/nrm2815] [Citation(s) in RCA: 350] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The regulated migration of cells is essential for development and tissue homeostasis, and aberrant cell migration can lead to an impaired immune response and the progression of cancer. Primordial germ cells (PGCs), precursors to sperm and eggs, have to migrate across the embryo to reach somatic gonadal precursors, where they carry out their function. Studies of model organisms have revealed that, despite important differences, several features of PGC migration are conserved. PGCs require an intrinsic motility programme and external guidance cues to survive and successfully migrate. Proper guidance involves both attractive and repulsive cues and is mediated by protein and lipid signalling.
Collapse
Affiliation(s)
- Brian E Richardson
- Howard Hughes Medical Institute, Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York University, New York, 10016, USA
| | | |
Collapse
|
24
|
Ruiz-Vela A, Aguilar-Gallardo C, Simón C. Building a Framework for Embryonic Microenvironments and Cancer Stem Cells. Stem Cell Rev Rep 2009; 5:319-27. [DOI: 10.1007/s12015-009-9096-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
25
|
Sheng XR, Brawley CM, Matunis EL. Dedifferentiating spermatogonia outcompete somatic stem cells for niche occupancy in the Drosophila testis. Cell Stem Cell 2009; 5:191-203. [PMID: 19664993 PMCID: PMC2750779 DOI: 10.1016/j.stem.2009.05.024] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 04/24/2009] [Accepted: 05/28/2009] [Indexed: 01/15/2023]
Abstract
Differentiating cells can dedifferentiate to replace stem cells in aged or damaged tissues, but the underlying mechanisms are unknown. In the Drosophila testis, a cluster of stromal cells called the hub creates a niche by locally activating Janus kinase-signal transducer and activator of transcription (Jak-STAT) signaling in adjacent germline and somatic stem cells. Here, we establish a system to study spermatogonial dedifferentiation. Ectopically expressing the differentiation factor bag-of-marbles (Bam) removes germline stem cells from the niche. However, withdrawing ectopic Bam causes interconnected spermatogonia to fragment, move into the niche, exchange positions with resident somatic stem cells, and establish contact with the hub. Concomitantly, actin-based protrusions appear on subsets of spermatogonia, suggesting acquired motility. Furthermore, global downregulation of Jak-STAT signaling inhibits dedifferentiation, indicating that normal levels of pathway activation are required to promote movement of spermatogonia into the niche during dedifferentiation, where they outcompete somatic stem cells for niche occupancy.
Collapse
Affiliation(s)
- X. Rebecca Sheng
- Department of Cell Biology, 725 N. Wolfe Street, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Crista M. Brawley
- Department of Cell Biology, 725 N. Wolfe Street, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Erika L. Matunis
- Department of Cell Biology, 725 N. Wolfe Street, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
26
|
Li WX. Canonical and non-canonical JAK-STAT signaling. Trends Cell Biol 2008; 18:545-51. [PMID: 18848449 PMCID: PMC3082280 DOI: 10.1016/j.tcb.2008.08.008] [Citation(s) in RCA: 224] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2008] [Revised: 08/07/2008] [Accepted: 08/12/2008] [Indexed: 12/11/2022]
Abstract
Aberrant activation of the JAK-STAT pathway has been implicated in many human cancers. It has widely been assumed that the effects of STAT activation are mediated by direct transcriptional induction of STAT target genes. However, recent findings in Drosophila have identified a non-canonical mode of JAK-STAT signaling, which directly controls heterochromatin stability. This indicates that the JAK-STAT pathway also controls cellular epigenetic status, which affects expression of genes beyond those under direct STAT transcriptional control. Given the evolutionary conservation of the canonical pathway among different species, the non-canonical mode of JAK-STAT signaling might also operate in vertebrates. In this review, canonical versus non-canonical JAK-STAT signaling and the implications for gene regulation and cancer formation are discussed.
Collapse
Affiliation(s)
- Willis X Li
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Avenue, KMRB 2-9641, Rochester, NY 14642, USA.
| |
Collapse
|
27
|
Gregory L, Came PJ, Brown S. Stem cell regulation by JAK/STAT signaling in Drosophila. Semin Cell Dev Biol 2008; 19:407-13. [PMID: 18603010 DOI: 10.1016/j.semcdb.2008.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 06/12/2008] [Indexed: 11/16/2022]
Abstract
Stem cells have become one of the "buzz" topics in the last decade or so. One of the best systems to study adult stem cells in vivo is in the model organism, Drosophila melanogaster. One hundred years of genetic analysis, a sequenced and highly annotated genome and genomics makes this a difficult organism to avoid. The JAK/STAT pathway has been shown to regulate stem cells during haematopoiesis and gametogenesis in Drosophila. In this review we cover the current literature and contrast each group of stem cells with respect to JAK/STAT signaling.
Collapse
Affiliation(s)
- Lorna Gregory
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | | | | |
Collapse
|
28
|
Disclosing JAK/STAT links to cell adhesion and cell polarity. Semin Cell Dev Biol 2008; 19:370-8. [DOI: 10.1016/j.semcdb.2008.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 06/05/2008] [Accepted: 06/06/2008] [Indexed: 12/27/2022]
|
29
|
Pontoriero GF, Deschamps P, Ashery-Padan R, Wong R, Yang Y, Zavadil J, Cvekl A, Sullivan S, Williams T, West-Mays JA. Cell autonomous roles for AP-2alpha in lens vesicle separation and maintenance of the lens epithelial cell phenotype. Dev Dyn 2008; 237:602-17. [PMID: 18224708 PMCID: PMC2517426 DOI: 10.1002/dvdy.21445] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In this study, we have created a conditional deletion of AP-2alpha in the developing mouse lens (Le-AP-2alpha mutants) to determine the cell-autonomous requirement(s) for AP-2alpha in lens development. Embryonic and adult Le-AP-2alpha mutants exhibited defects confined to lens placode derivatives, including a persistent adhesion of the lens to the overlying corneal epithelium (or lens stalk). Expression of known regulators of lens vesicle separation, including Pax6, Pitx3, and Foxe3 was observed in the Le-AP-2alpha mutant lens demonstrating that these genes do not lie directly downstream of AP-2alpha. Unlike germ-line mutants, Le-AP-2alpha mutants did not exhibit defects in the optic cup, further defining the tissue specific role(s) for AP-2alpha in eye development. Finally, comparative microarray analysis of lenses from the Le-AP-2alpha mutants vs. wild-type littermates revealed differential expression of 415 mRNAs, including reduced expression of genes important for maintaining the lens epithelial cell phenotype, such as E-cadherin.
Collapse
Affiliation(s)
- Giuseppe F. Pontoriero
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Paula Deschamps
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ruth Ashery-Padan
- Human Genetics and Molecular Medicine Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Ryan Wong
- Departments of Ophthalmology and Visual Sciences and Molecular Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Ying Yang
- Departments of Ophthalmology and Visual Sciences and Molecular Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Jiri Zavadil
- Department of Pathology and NYU Cancer Institute, NYU School of Medicine, New York, New York
| | - Ales Cvekl
- Departments of Ophthalmology and Visual Sciences and Molecular Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Shelley Sullivan
- Departments of CFB and CDB, University of Colorado Health Sciences Center, Denver, Colorado
| | - Trevor Williams
- Departments of CFB and CDB, University of Colorado Health Sciences Center, Denver, Colorado
| | - Judith A. West-Mays
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
30
|
Abstract
The basic concepts of the molecular machinery that mediates cell migration have been gleaned from cell culture systems. However, the three-dimensional environment within an organism presents migrating cells with a much greater challenge. They must move between and among other cells while interpreting multiple attractive and repulsive cues to choose their proper path. They must coordinate their cell adhesion with their surroundings and know when to start and stop moving. New insights into the control of these remaining mysteries have emerged from genetic dissection and live imaging of germ cell migration in Drosophila, zebrafish, and mouse embryos. In this review, we first describe germ cell migration in cellular and mechanistic detail in these different model systems. We then compare these systems to highlight the emerging principles. Finally, we contrast the migration of germ cells with that of immune and cancer cells to outline the conserved and different mechanisms.
Collapse
Affiliation(s)
- Prabhat S Kunwar
- Howard Hughes Medical Institute, Developmental Genetics Program, Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York, New York 10016-6402, USA.
| | | | | |
Collapse
|
31
|
Arbouzova NI, Zeidler MP. JAK/STAT signalling in Drosophila: insights into conserved regulatory and cellular functions. Development 2006; 133:2605-16. [PMID: 16794031 DOI: 10.1242/dev.02411] [Citation(s) in RCA: 306] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
High levels of interspecies conservation characterise all signal transduction cascades and demonstrate the significance of these pathways over evolutionary time. Here, we review advances in the field of JAK/STAT signalling, focusing on recent developments in Drosophila. In particular, recent results from genetic and genome-wide RNAi screens, as well as studies into the developmental roles played by this pathway, highlight striking levels of physical and functional conservation in processes such as cellular proliferation, immune responses and stem cell maintenance. These insights underscore the value of model organisms for improving our understanding of this human disease-relevant pathway.
Collapse
Affiliation(s)
- Natalia I Arbouzova
- Department of Molecular Developmental Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | | |
Collapse
|