Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 26(6): 859-867, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38689013

ABSTRACT

Tissue regeneration and maintenance rely on coordinated stem cell behaviours. This orchestration can be impaired by oncogenic mutations leading to cancer. However, it is largely unclear how oncogenes perturb stem cells' orchestration to disrupt tissue. Here we used intravital imaging to investigate the mechanisms by which oncogenic Kras mutation causes tissue disruption in the hair follicle. Through longitudinally tracking hair follicles in live mice, we found that KrasG12D, a mutation that can lead to squamous cell carcinoma, induces epithelial tissue deformation in a spatiotemporally specific manner, linked with abnormal cell division and migration. Using a reporter mouse capture real-time ERK signal dynamics at the single-cell level, we discovered that KrasG12D, but not a closely related mutation HrasG12V, converts ERK signal in stem cells from pulsatile to sustained. Finally, we demonstrated that interrupting sustained ERK signal reverts KrasG12D-induced tissue deformation through modulating specific features of cell migration and division.


Subject(s)
Cell Movement , Hair Follicle , Mutation , Proto-Oncogene Proteins p21(ras) , Animals , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mice , Hair Follicle/metabolism , MAP Kinase Signaling System/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Mice, Transgenic , Stem Cells/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Humans , Female , Enzyme Activation
2.
Cell Stem Cell ; 30(7): 962-972.e6, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37419106

ABSTRACT

The ability of stem cells to build and replenish tissues depends on support from their niche. Although niche architecture varies across organs, its functional importance is unclear. During hair follicle growth, multipotent epithelial progenitors build hair via crosstalk with their remodeling fibroblast niche, the dermal papilla, providing a powerful model to functionally interrogate niche architecture. Through mouse intravital imaging, we show that dermal papilla fibroblasts remodel individually and collectively to form a morphologically polarized, structurally robust niche. Asymmetric TGF-ß signaling precedes morphological niche polarity, and loss of TGF-ß signaling in dermal papilla fibroblasts leads them to progressively lose their stereotypic architecture, instead surrounding the epithelium. The reorganized niche induces the redistribution of multipotent progenitors but nevertheless supports their proliferation and differentiation. However, the differentiated lineages and hairs produced by progenitors are shorter. Overall, our results reveal that niche architecture optimizes organ efficiency but is not absolutely essential for organ function.


Subject(s)
Hair Follicle , Hair , Mice , Animals , Cell Differentiation , Epithelium , Transforming Growth Factor beta
3.
Nature ; 619(7968): 167-175, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37344586

ABSTRACT

Healthy skin is a mosaic of wild-type and mutant clones1,2. Although injury can cooperate with mutated Ras family proteins to promote tumorigenesis3-12, the consequences in genetically mosaic skin are unknown. Here we show that after injury, wild-type cells suppress aberrant growth induced by oncogenic Ras. HrasG12V/+ and KrasG12D/+ cells outcompete wild-type cells in uninjured, mosaic tissue but their expansion is prevented after injury owing to an increase in the fraction of proliferating wild-type cells. Mechanistically, we show that, unlike HrasG12V/+ cells, wild-type cells respond to autocrine and paracrine secretion of EGFR ligands, and this differential activation of the EGFR pathway explains the competitive switch during injury repair. Inhibition of EGFR signalling via drug or genetic approaches diminishes the proportion of dividing wild-type cells after injury, leading to the expansion of HrasG12V/+ cells. Increased proliferation of wild-type cells via constitutive loss of the cell cycle inhibitor p21 counteracts the expansion of HrasG12V/+ cells even in the absence of injury. Thus, injury has a role in switching the competitive balance between oncogenic and wild-type cells in genetically mosaic skin.


Subject(s)
Cell Proliferation , Genes, ras , Mosaicism , Mutation , Skin , ras Proteins , Cell Cycle , Cell Proliferation/genetics , ErbB Receptors/metabolism , ras Proteins/genetics , ras Proteins/metabolism , Skin/cytology , Skin/injuries , Skin/metabolism , Skin/pathology , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism
4.
Elife ; 122023 03 07.
Article in English | MEDLINE | ID: mdl-36880644

ABSTRACT

Stem cell differentiation requires dramatic changes in gene expression and global remodeling of chromatin architecture. How and when chromatin remodels relative to the transcriptional, behavioral, and morphological changes during differentiation remain unclear, particularly in an intact tissue context. Here, we develop a quantitative pipeline which leverages fluorescently-tagged histones and longitudinal imaging to track large-scale chromatin compaction changes within individual cells in a live mouse. Applying this pipeline to epidermal stem cells, we reveal that cell-to-cell chromatin compaction heterogeneity within the stem cell compartment emerges independent of cell cycle status, and instead is reflective of differentiation status. Chromatin compaction state gradually transitions over days as differentiating cells exit the stem cell compartment. Moreover, establishing live imaging of Keratin-10 (K10) nascent RNA, which marks the onset of stem cell differentiation, we find that Keratin-10 transcription is highly dynamic and largely precedes the global chromatin compaction changes associated with differentiation. Together, these analyses reveal that stem cell differentiation involves dynamic transcriptional states and gradual chromatin rearrangement.


Subject(s)
Chromatin , Keratin-10 , Animals , Mice , Keratin-10/genetics , Keratin-10/metabolism , Histones/metabolism , Cell Differentiation/genetics , Stem Cells/metabolism
5.
Dev Cell ; 55(3): 328-340.e5, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33091369

ABSTRACT

Despite the noisy nature of single cells, multicellular organisms robustly generate different cell types from one zygote. This process involves dynamic cross regulation between signaling and gene expression that is difficult to capture with fixed-cell approaches. To study signaling dynamics and fate specification during preimplantation development, we generated a transgenic mouse expressing the ERK kinase translocation reporter and measured ERK activity in single cells of live embryos. Our results show primarily active ERK in both the inner cell mass and trophectoderm cells due to fibroblast growth factor (FGF) signaling. Strikingly, a subset of mitotic events results in a short pulse of ERK inactivity in both daughter cells that correlates with elevated endpoint NANOG levels. Moreover, endogenous tagging of Nanog in embryonic stem cells reveals that ERK inhibition promotes enhanced stabilization of NANOG protein after mitosis. Our data show that cell cycle, signaling, and differentiation are coordinated during preimplantation development.


Subject(s)
Blastocyst/cytology , Blastocyst/enzymology , Cell Cycle , Cell Lineage , MAP Kinase Signaling System , Mammals/embryology , Animals , Germ Layers/cytology , Humans , Mice , Mitosis , Models, Biological , Nanog Homeobox Protein/metabolism , Protein Stability , Reproducibility of Results
6.
J Cell Biol ; 218(10): 3212-3222, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31488583

ABSTRACT

Mutations associated with tumor development in certain tissues can be nontumorigenic in others, yet the mechanisms underlying these different outcomes remains poorly understood. To address this, we targeted an activating Hras mutation to hair follicle stem cells and discovered that Hras mutant cells outcompete wild-type neighbors yet are integrated into clinically normal skin hair follicles. In contrast, targeting the Hras mutation to the upper noncycling region of the skin epithelium leads to benign outgrowths. Follicular Hras mutant cells autonomously and nonautonomously enhance regeneration, which directs mutant cells into continuous tissue cycling to promote integration rather than aberrancy. This follicular tolerance is maintained under additional challenges that promote tumorigenesis in the epidermis, including aging, injury, and a secondary mutation. Thus, the hair follicle possesses a unique, enhanced capacity to integrate and contain Hras mutant cells within both homeostatic and perturbed tissue, demonstrating that in the skin, multiple, distinct mechanisms exist to suppress oncogenic growth.


Subject(s)
Carcinogenesis , Hair Follicle/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Regeneration , ras Proteins/metabolism , Animals , Mice , Mice, Transgenic
7.
Nat Cell Biol ; 20(12): 1361-1369, 2018 12.
Article in English | MEDLINE | ID: mdl-30420661

ABSTRACT

Tissue homeostasis is sustained by stem cell self-renewal and differentiation. How stem cells coordinately differentiate into multiple cell types is largely unclear. Recent studies underline the heterogeneity among stem cells or common progenitors, suggesting that coordination occurs at the stem cell/progenitor level1-4. Here, by tracking and manipulating the same stem cells and their progeny at the single-cell level in live mice, we uncover an unanticipated flexibility of homeostatic stem cell differentiation in hair follicles. Although stem cells have been shown to be flexible upon injury, we demonstrate that hair germ stem cells at the single-cell level can flexibly establish all of the differentiation lineages even in uninjured conditions. Furthermore, stem cell-derived hair progenitors in the structure called matrix, previously thought to be unipotent, flexibly change differentiation outcomes as a consequence of unexpected dynamic relocation. Finally, the flexible cell fate determination mechanism maintains normal differentiation and tissue architecture against an ectopic differentiation stimulus induced by Wnt activation. This work provides a model of continual fate channelling and late commitment of stem cells to achieve coordinated differentiation and robust tissue architecture.


Subject(s)
Cell Differentiation , Cell Lineage , Hair Follicle/cytology , Stem Cells/cytology , Animals , Hair Follicle/metabolism , Homeostasis , Mice, Knockout , Mice, Transgenic , Single-Cell Analysis/methods , Stem Cells/metabolism
8.
Cell Stem Cell ; 23(5): 677-686.e4, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30269903

ABSTRACT

Maintenance of adult tissues depends on sustained activity of resident stem cell populations, but the mechanisms that regulate stem cell self-renewal during homeostasis remain largely unknown. Using an imaging and tracking approach that captures all epidermal stem cell activity in large regions of living mice, we show that self-renewal is locally coordinated with epidermal differentiation, with a lag time of 1 to 2 days. In both homeostasis and upon experimental perturbation, we find that differentiation of a single stem cell is followed by division of a direct neighbor, but not vice versa. Finally, we show that exit from the stem cell compartment is sufficient to drive neighboring stem cell self-renewal. Together, these findings establish that epidermal stem cell self-renewal is not the constitutive driver of homeostasis. Instead, it is precisely tuned to tissue demand and responds directly to neighbor cell differentiation.


Subject(s)
Cell Differentiation , Epidermal Cells/cytology , Homeostasis , Stem Cells/cytology , Animals , Epidermal Cells/metabolism , Epidermis/metabolism , Female , Male , Mice , Stem Cells/metabolism
9.
Nature ; 548(7667): 334-337, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28783732

ABSTRACT

Cells in healthy tissues acquire mutations with surprising frequency. Many of these mutations are associated with abnormal cellular behaviours such as differentiation defects and hyperproliferation, yet fail to produce macroscopically detectable phenotypes. It is currently unclear how the tissue remains phenotypically normal, despite the presence of these mutant cells. Here we use intravital imaging to track the fate of mouse skin epithelium burdened with varying numbers of activated Wnt/ß-catenin stem cells. We show that all resulting growths that deform the skin tissue architecture regress, irrespective of their size. Wild-type cells are required for the active elimination of mutant cells from the tissue, while utilizing both endogenous and ectopic cellular behaviours to dismantle the aberrant structures. After regression, the remaining structures are either completely eliminated or converted into functional skin appendages in a niche-dependent manner. Furthermore, tissue aberrancies generated from oncogenic Hras, and even mutation-independent deformations to the tissue, can also be corrected, indicating that this tolerance phenomenon reflects a conserved principle in the skin. This study reveals an unanticipated plasticity of the adult skin epithelium when faced with mutational and non-mutational insult, and elucidates the dynamic cellular behaviours used for its return to a homeostatic state.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Homeostasis , Mutation , Phenotype , Skin/cytology , Animals , Mice , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
10.
Dev Biol ; 416(2): 312-23, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27339292

ABSTRACT

The Drosophila larval ovary morphogenesis mainly involves coordinated development of somatic and germ cell lineages that is essential for forming a correct number of niche-germline stem cell (GSC) units (ovarioles) in the adult ovary. Ecdysone, Insulin, Activin, Dpp and EGFR signaling pathways form a regulatory network that orchestrates ovarian soma and germ line throughout larval development. Identification and characterization of additional genes or machineries involved in this process will provide more insights into the underlying mechanisms. Here, we show that the core microRNA (miRNA) pathway components Drosha and Pasha are required for coordinated development of somatic and germ cell precursors in the larval ovary. Drosha or pasha mutants display defective proliferation of primordial germ cells (PGCs), the precursors of GSCs prior to late third larval instar (LL3) and promoted PGC differentiation at LL3. In the mean time, loss of Drosha or Pasha function perturbs somatic precursor development, causing defects in formation of terminal filaments (TFs), a major composition of the GSC niche at LL3, as well as in TF precursor accumulation at early larval stages. Comparative analysis of the mutant phenotypes reveals that three other key miRNA pathway components, Dicer-1 (Dcr-1), Loquacious (Loqs) and Argonaute-1 (Ago-1) have similar effects as Drosha and Pasha indicated above, suggesting a role of the canonical miRNA pathway in the ovary development. Furthermore, genome-wide screening and genetic studies identify a set of Drosha-controlled miRNAs including miR-8, miR-14, miR-33, miR-184, miR-317 and let-7-C that function in this gonadogenesis. Taken together, this study provides the first ever demonstration that miRNA-mediated regulation is involved in the Drosophila larval ovary morphogenesis.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/genetics , MicroRNAs/genetics , Ovary/growth & development , RNA-Binding Proteins/physiology , Ribonuclease III/physiology , Animals , Cell Differentiation , Cytoskeleton/ultrastructure , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Embryonic Germ Cells/cytology , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Larva , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Microscopy, Fluorescence , Organogenesis , Ovary/cytology , RNA Interference , RNA-Binding Proteins/genetics , Ribonuclease III/deficiency , Ribonuclease III/genetics , Stem Cell Niche
11.
Cell ; 164(6): 1212-1225, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26967287

ABSTRACT

Adult stem cells across diverse organs self-renew and differentiate to maintain tissue homeostasis. How stem cells receive input to preserve tissue structure and function largely relies on their communication with surrounding cellular and non-cellular elements. As such, how tissues are organized and patterned not only reflects organ function, but also inherently hardwires networks of communication between stem cells and their environment to direct tissue homeostasis and injury repair. This review highlights how different methods of stem cell communication reflect the unique organization and function of diverse tissues.


Subject(s)
Cell Communication , Adult Stem Cells/cytology , Animals , Homeostasis , Humans , Regeneration , Stem Cells/cytology
12.
PLoS One ; 9(7): e103473, 2014.
Article in English | MEDLINE | ID: mdl-25068272

ABSTRACT

In the Drosophila oogenesis, germline stem cells (GSCs) continuously self-renew and differentiate into daughter cells for consecutive germline lineage commitment. This developmental process has become an in vivo working platform for studying adult stem cell fate regulation. An increasing number of studies have shown that while concerted actions of extrinsic signals from the niche and intrinsic regulatory machineries control GSC self-renewal and germline differentiation, epigenetic regulation is implicated in the process. Here, we report that Brahma (Brm), the ATPase subunit of the Drosophila SWI/SNF chromatin-remodeling complexes, is required for maintaining GSC fate. Removal or knockdown of Brm function in either germline or niche cells causes a GSC loss, but does not disrupt normal germline differentiation within the germarium evidenced at the molecular and morphological levels. There are two Drosophila SWI/SNF complexes: the Brm-associated protein (BAP) complex and the polybromo-containing BAP (PBAP) complex. More genetic studies reveal that mutations in polybromo/bap180, rather than gene encoding Osa, the BAP complex-specific subunit, elicit a defect in GSC maintenance reminiscent of the brm mutant phenotype. Further genetic interaction test suggests a functional association between brm and polybromo in controlling GSC self-renewal. Taken together, studies in this paper provide the first demonstration that Brm in the form of the PBAP complex functions in the GSC fate regulation.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Drosophila Proteins/genetics , Ovum/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microscopy, Confocal , Mutation , Ovary/cytology , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/metabolism , Transcription Factors/metabolism
13.
Dev Biol ; 384(1): 1-12, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24120347

ABSTRACT

Maintenance of adult stem cells is largely dependent on the balance between their self-renewal and differentiation. The Drosophila ovarian germline stem cells (GSCs) provide a powerful in vivo system for studying stem cell fate regulation. It has been shown that maintaining the GSC population involves both genetic and epigenetic mechanisms. Although the role of epigenetic regulation in this process is evident, the underlying mechanisms remain to be further explored. In this study, we find that Enoki mushroom (Enok), a Drosophila putative MYST family histone acetyltransferase controls GSC maintenance in the ovary at multiple levels. Removal or knockdown of Enok in the germline causes a GSC maintenance defect. Further studies show that the cell-autonomous role of Enok in maintaining GSCs is not dependent on the BMP/Bam pathway. Interestingly, molecular studies reveal an ectopic expression of Bruno, an RNA binding protein, in the GSCs and their differentiating daughter cells elicited by the germline Enok deficiency. Misexpression of Bruno in GSCs and their immediate descendants results in a GSC loss that can be exacerbated by incorporating one copy of enok mutant allele. These data suggest a role for Bruno in Enok-controlled GSC maintenance. In addition, we observe that Enok is required for maintaining GSCs non-autonomously. Compromised expression of enok in the niche cells impairs the niche maintenance and BMP signal output, thereby causing defective GSC maintenance. This is the first demonstration that the niche size control requires an epigenetic mechanism. Taken together, studies in this paper provide new insights into the GSC fate regulation.


Subject(s)
Drosophila Proteins/genetics , Drosophila/embryology , Drosophila/genetics , Germ Cells/metabolism , Histone Acetyltransferases/genetics , RNA-Binding Proteins/genetics , Stem Cell Niche/physiology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Drosophila/metabolism , Drosophila Proteins/metabolism , Embryo, Nonmammalian/metabolism , Epigenomics , Female , Histone Acetyltransferases/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/genetics
14.
Dev Biol ; 379(2): 167-81, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23624310

ABSTRACT

The Drosophila ovarian germline stem cells (GSCs) constantly experience self-renewal and differentiation, ensuring the female fertility throughout life. The balance between GSC self-renewal and differentiation is exquisitely regulated by the stem cell niche, the stem cells themselves and systemic factors. Increasing evidence has shown that the GSC regulation also involves epigenetic mechanisms including chromatin remodeling and histone modification. Here, we find that dBre1, an E3 ubiquitin ligase, functions in controlling GSC self-renewal and germ cell differentiation via distinct mechanisms. Removal or knock down of dBre1 function in the germline or somatic niche cell lineage leads to a gradual GSC loss and disruption of H3K4 trimethylation in the Drosophila ovary. Further studies suggest that the defective GSC maintenance is attributable to compromised BMP signaling emitted from the stem cell niche and impaired adhesion of GSCs to their niche. On the other hand, dBre1-RNAi expression in escort cells causes a loss of H3K4 trimethylation and accumulation of spectrosome-containing single germ cells in the germarium. Reducing dpp or dally levels suppresses the germ cell differentiation defects, indicating that dBre1 limits BMP signaling activities for the differentiation control. Strikingly, all phenotypes observed in dBre1 mutant ovaries can be mimicked by RNAi-based reduced expression of dSet1, a Drosophila H3K4 trimethylase. Moreover, genetic studies favor that dBre1 interacts with dSet1 in controlling GSC maintenance and germ cell differentiation. Taken together, we identify a dBre1/dSet1-dependent pathway for the H3K4 methylation involved in the cell fate regulation in the Drosophila ovary.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Differentiation/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Germ Cells/physiology , Ovary/cytology , Ubiquitin-Protein Ligases/metabolism , Animals , DNA Primers/genetics , Epigenesis, Genetic/physiology , Female , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methylation , Microscopy, Fluorescence , Ovary/embryology , RNA Interference , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric , Stem Cell Niche/physiology , Stem Cells/physiology , Ubiquitin-Protein Ligases/genetics
16.
BMC Dev Biol ; 9: 60, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19948068

ABSTRACT

BACKGROUND: Proper patterning of the follicle cell epithelium over the egg chamber is essential for the Drosophila egg development. Differentiation of the epithelium into several distinct cell types along the anterior-posterior axis requires coordinated activities of multiple signaling pathways. Previously, we reported that lethal(2)giant larvae (lgl), a Drosophila tumor suppressor gene, is required in the follicle cells for the posterior follicle cell (PFC) fate induction at mid-oogenesis. Here we explore the role of another two tumor suppressor genes, scribble (scrib) and discs large (dlg), in the epithelial patterning. RESULTS: We found that removal of scrib or dlg function from the follicle cells at posterior terminal of the egg chamber causes a complete loss of the PFC fate. Aberrant specification and differentiation of the PFCs in the mosaic clones can be ascribed to defects in coordinated activation of the EGFR, JAK and Notch signaling pathways in the multilayered cells. Meanwhile, the clonal analysis revealed that loss-of-function mutations in scrib/dlg at the anterior domains result in a partially penetrant phenotype of defective induction of the stretched and centripetal cell fate, whereas specification of the border cell fate can still occur in the most anterior region of the mutant clones. Further, we showed that scrib genetically interacts with dlg in regulating posterior patterning of the epithelium. CONCLUSION: In this study we provide evidence that scrib and dlg function differentially in anterior and posterior patterning of the follicular epithelium at oogenesis. Further genetic analysis indicates that scrib and dlg act in a common pathway to regulate PFC fate induction. This study may open another window for elucidating role of scrib/dlg in controlling epithelial polarity and cell proliferation during development.


Subject(s)
Drosophila Proteins/physiology , Epithelium/embryology , Epithelium/metabolism , Membrane Proteins/physiology , Oogenesis/physiology , Ovarian Follicle/embryology , Ovarian Follicle/metabolism , Tumor Suppressor Proteins/physiology , Animals , Body Patterning/genetics , Body Patterning/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Drosophila , Drosophila Proteins/genetics , Female , Membrane Proteins/genetics , Oogenesis/genetics , Ovarian Follicle/cytology , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics
17.
Cell Res ; 18(3): 372-84, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18268543

ABSTRACT

The intricately regulated differentiation of the somatic follicle cell lineages into distinct subpopulations with specific functions plays an essential role in Drosophila egg development. At early oogenesis, induction of the stalk cells generates the first anteroposterior (AP) asymmetry in the egg chamber by inducing the posterior localization of the oocyte. Later, the properly specified posterior follicle cells signal to polarize the oocyte along the AP and dorsoventral (DV) axes at mid-oogenesis. Here, we show that lethal(2)giant larvae (lgl), a Drosophila tumor suppressor gene, is required in the follicle cells for the differentiation of both stalk cells and posterior follicle cells. Loss-of-function mutations in lgl cause oocyte mispositioning in the younger one of the fused chambers, due to lack of the stalk. Removal of lgl function from the posterior follicle cells using the FLP/FRT system results in loss of the oocyte polarity that is elicited by the failure of those posterior cells to differentiate normally. Thus, we provide the first demonstration that lgl is implicated in the formation of the initial AP asymmetry and the patterning of the AP and DV axes in the oocyte by acting in the specification of a subset of somatic follicle cells.


Subject(s)
Cell Polarity/physiology , Drosophila Proteins/metabolism , Oocytes/metabolism , Oogenesis/physiology , Tumor Suppressor Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Male , Mutation , Oocytes/cytology , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...