Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Biol ; 31(21): 4697-4712.e6, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34480855

ABSTRACT

Stem cell homeostasis requires coordinated fate decisions among stem cells that are often widely distributed within a tissue at varying distances from their stem cell niche. This requires a mechanism to ensure robust fate decisions within a population of stem cells. Here, we show that, in the Drosophila hematopoietic organ, the lymph gland (LG), gap junctions form a network that coordinates fate decisions between blood progenitors. Using live imaging of calcium signaling in intact LGs, we find that blood progenitors are connected through a signaling network. Blocking gap junction function disrupts this network, alters the pattern of encoded calcium signals, and leads to loss of progenitors and precocious blood cell differentiation. Ectopic and uniform activation of the calcium-signaling mediator CaMKII restores progenitor homeostasis when gap junctions are disrupted. Overall, these data show that gap junctions equilibrate cell signals between blood progenitors to coordinate fate decisions and maintain hematopoietic homeostasis.


Subject(s)
Calcium , Drosophila Proteins , Animals , Calcium Signaling , Cell Differentiation/physiology , Drosophila/physiology , Drosophila Proteins/metabolism , Gap Junctions/metabolism , Hematopoiesis/physiology
2.
Curr Biol ; 30(17): 3316-3329.e5, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32649911

ABSTRACT

During hematopoiesis, progenitor cells receive and interpret a diverse array of regulatory signals from their environment. These signals control the maintenance of the progenitors and regulate the production of mature blood cells. Integrins are well known in vertebrates for their roles in hematopoiesis, particularly in assisting in the migration to, as well as the physical attachment of, progenitors to the niche. However, whether and how integrins are also involved in the signaling mechanisms that control hematopoiesis remains to be resolved. Here, we show that integrins play a key role during fly hematopoiesis in regulating cell signals that control the behavior of hematopoietic progenitors. Integrins can regulate hematopoiesis directly, via focal adhesion kinase (FAK) signaling, and indirectly, by directing extracellular matrix (ECM) assembly and/or maintenance. ECM organization and density controls blood progenitor behavior by modulating multiple signaling pathways, including bone morphogenetic protein (BMP) and Hedgehog (Hh). Furthermore, we show that integrins and the ECM are reduced following infection, which may assist in activating the immune response. Our results provide mechanistic insight into how integrins can shape the signaling environment around hematopoietic progenitors.


Subject(s)
Blood Cells/immunology , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Extracellular Matrix/physiology , Hematopoiesis , Integrins/metabolism , Animals , Blood Cells/metabolism , Blood Cells/parasitology , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/parasitology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Integrins/genetics , Signal Transduction , Wasps/physiology
3.
Development ; 146(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30890573

ABSTRACT

Hematopoiesis requires coordinated cell signals to control the proliferation and differentiation of progenitor cells. In Drosophila, blood progenitors, called prohemocytes, which are located in a hematopoietic organ called the lymph gland, are regulated by the Salvador-Warts-Hippo pathway. In epithelial cells, the Hippo pathway integrates diverse biological inputs, such as cell polarity and cell-cell contacts, but Drosophila blood cells lack the conspicuous polarity of epithelial cells. Here, we show that the septate-junction components Cora and NrxIV promote Hippo signaling in the lymph gland. Depletion of septate-junction components in hemocytes produces similar phenotypes to those observed in Hippo pathway mutants, including increased differentiation of immune cells. Our analysis places septate-junction components as upstream regulators of the Hippo pathway where they recruit Merlin to the membrane. Finally, we show that interactions of septate-junction components with the Hippo pathway are a key functional component of the cellular immune response following infection.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Tight Junctions/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Polarity/genetics , Cell Polarity/physiology , Drosophila Proteins/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hematopoiesis/genetics , Hematopoiesis/physiology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/metabolism , Tight Junctions/genetics
4.
Elife ; 62017 08 25.
Article in English | MEDLINE | ID: mdl-28841136

ABSTRACT

Stem cells are regulated by signals from their microenvironment, or niche. During Drosophila hematopoiesis, a niche regulates prohemocytes to control hemocyte production. Immune challenges activate cell-signalling to initiate the cellular and innate immune response. Specifically, certain immune challenges stimulate the niche to produce signals that induce prohemocyte differentiation. However, the mechanisms that promote prohemocyte differentiation subsequent to immune challenges are poorly understood. Here we show that bacterial infection induces the cellular immune response by modulating occluding-junctions at the hematopoietic niche. Occluding-junctions form a permeability barrier that regulates the accessibility of prohemocytes to niche derived signals. The immune response triggered by infection causes barrier breakdown, altering the prohemocyte microenvironment to induce immune cell production. Moreover, genetically induced barrier ablation provides protection against infection by activating the immune response. Our results reveal a novel role for occluding-junctions in regulating niche-hematopoietic progenitor signalling and link this mechanism to immune cell production following infection.


Subject(s)
Cell Differentiation , Drosophila/immunology , Hemocytes/immunology , Hemocytes/physiology , Tight Junctions/metabolism , Animals , Bacteria/immunology
5.
Sci Rep ; 7(1): 118, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28273919

ABSTRACT

How multicellular organisms maintain immune homeostasis across various organs and cell types is an outstanding question in immune biology and cell signaling. In Drosophila, blood cells (hemocytes) respond to local and systemic cues to mount an immune response. While endosomal regulation of Drosophila hematopoiesis is reported, the role of endosomal proteins in cellular and humoral immunity is not well-studied. Here we demonstrate a functional role for endosomal proteins in immune homeostasis. We show that the ubiquitous trafficking protein ADP Ribosylation Factor 1 (ARF1) and the hemocyte-specific endosomal regulator Asrij differentially regulate humoral immunity. Asrij and ARF1 play an important role in regulating the cellular immune response by controlling the crystal cell melanization and phenoloxidase activity. ARF1 and Asrij mutants show reduced survival and lifespan upon infection, indicating perturbed immune homeostasis. The ARF1-Asrij axis suppresses the Toll pathway anti-microbial peptides (AMPs) by regulating ubiquitination of the inhibitor Cactus. The Imd pathway is inversely regulated- while ARF1 suppresses AMPs, Asrij is essential for AMP production. Several immune mutants have reduced Asrij expression, suggesting that Asrij co-ordinates with these pathways to regulate the immune response. Our study highlights the role of endosomal proteins in modulating the immune response by maintaining the balance of AMP production. Similar mechanisms can now be tested in mammalian hematopoiesis and immunity.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Drosophila Proteins/metabolism , Drosophila/immunology , Endosomes/metabolism , Membrane Proteins/metabolism , ADP-Ribosylation Factor 1/genetics , Adenosine Monophosphate/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Drosophila/metabolism , Drosophila Proteins/genetics , Immunity, Cellular , Immunity, Humoral , Melanins/metabolism , Membrane Proteins/genetics , Monophenol Monooxygenase/metabolism , Mutation , Phosphoproteins/metabolism , Ubiquitination
6.
Proc Natl Acad Sci U S A ; 111(13): 4898-903, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24707047

ABSTRACT

Drosophila melanogaster larval hematopoiesis is a well-established model to study mechanisms that regulate hematopoietic niche maintenance and control of blood cell precursor (prohemocyte) differentiation. Molecules that perturb niche function affect the balance between prohemocytes and differentiated hemocytes. The conserved hemocyte-specific endosomal protein Asrij is essential for niche function and prohemocyte maintenance. Elucidating how subcellular trafficking molecules can regulate signaling presents an important challenge. Here we show that Asrij function is mediated by the Ras family GTPase Arf79F, the Drosophila homolog of ADP ribosylation factor 1 (ARF1), essential for clathrin coat assembly, Golgi architecture, and vesicular trafficking. ARF1 is expressed in the larval lymph gland and in circulating hemocytes and interacts with Asrij. ARF1-depleted lymph glands show loss of niche cells and prohemocyte maintenance with increased differentiation. Inhibiting ARF1 activation by knocking down its guanine nucleotide exchange factor (Gartenzwerg) or overexpressing its GTPAse-activating protein showed that ARF1-GTP is essential for regulating niche size and maintaining stemness. Activated ARF1 regulates Asrij levels in blood cells thereby mediating Asrij function. Asrij controls crystal cell differentiation by affecting Notch trafficking. ARF1 perturbation also leads to aberrant Notch trafficking and the Notch intracellular domain is stalled in sorting endosomes. Thus, ARF1 can regulate Drosophila blood cell homeostasis by regulating Asrij endocytic function. ARF1 also regulates signals arising from the niche and differentiated cells by integrating the insulin-mediated and PDGF-VEGF receptor signaling pathways. We propose that the conserved ARF1-Asrij endocytic axis modulates signals that govern hematopoietic development. Thus, Asrij affords tissue-specific control of global mechanisms involved in molecular traffic.


Subject(s)
Blood Cells/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Guanosine Triphosphate/metabolism , Homeostasis , Membrane Proteins/metabolism , Animals , Blood Cells/cytology , Cell Proliferation , Drosophila melanogaster/cytology , Hematopoiesis , Hemocytes/metabolism , Insulin/metabolism , Larva/cytology , Larva/metabolism , Lymph Nodes/metabolism , Phenotype , Protein Binding , Protein Transport , Receptors, Notch/metabolism , Signal Transduction
7.
Cell Rep ; 4(4): 649-58, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23972987

ABSTRACT

Asrij/OCIAD1 is an endosomal protein expressed in stem cells and cardiovascular lineages and aberrantly expressed in several cancers. We show that dose-dependent modulation of cytokine-dependent JAK/STAT signaling by Asrij regulates mouse embryonic stem cell pluripotency as well as Drosophila hematopoietic stem cell maintenance. Furthermore, mouse asrij can substitute for Drosophila asrij, indicating that they are true homologs. We identify a conserved region of Asrij that is necessary and sufficient for vesicular localization and function. We also show that Asrij and STAT3 colocalize in endosomes and interact biochemically. We propose that Asrij provides an endosomal scaffold for STAT3 interaction and activation, and may similarly control other circuits that maintain stemness. Thus, Asrij provides a key point of control for spatial and kinetic regulation of stem cell signals.


Subject(s)
Cell Differentiation , Drosophila Proteins/metabolism , F-Box Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Janus Kinases/metabolism , Membrane Proteins/metabolism , Pluripotent Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Proliferation , Conserved Sequence , Drosophila , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Endosomes/metabolism , F-Box Proteins/genetics , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Pluripotent Stem Cells/cytology , Protein Binding , Protein Transport , Signal Transduction
8.
PLoS One ; 6(11): e27667, 2011.
Article in English | MEDLINE | ID: mdl-22110713

ABSTRACT

Several signaling pathways control blood cell (hemocyte) development in the Drosophila lymph gland. Mechanisms that modulate and integrate these signals are poorly understood. Here we report that mutation in a conserved endocytic protein Asrij affects signal transmission and causes aberrant lymph gland hematopoiesis. Mammalian Asrij (Ociad1) is expressed in stem cells of the blood vascular system and is implicated in several cancers. We found that Drosophila Asrij is a pan-hemocyte marker and localizes to a subset of endocytic vesicles. Loss of asrij causes hyperproliferation of lymph gland lobes coupled with increased hemocyte differentiation, thereby depleting the pool of quiescent hemocyte precursors. This co-relates with fewer Col+ cells in the hematopoietic stem cell niche of asrij mutants. Asrij null mutants also show excess specification of crystal cells that express the RUNX factor Lozenge (Lz), a target of Notch signaling. Asrij mutant lymph glands show increased N in sorting endosomes suggesting aberrant trafficking. In vitro assays also show impaired traffic of fluorescent probes in asrij null hemocytes. Taken together our data suggest a role for Asrij in causing increased Notch signaling thereby affecting hemocyte differentiation. Thus, conserved endocytic functions may control blood cell progenitor quiescence and differentiation.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Hematopoiesis, Extramedullary , Lymph Nodes/cytology , Membrane Proteins/metabolism , Stem Cell Niche , Animals , Biomarkers/metabolism , Cell Count , Cell Proliferation , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/physiology , Endocytosis , Endosomes/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Hemocytes/cytology , Hemocytes/metabolism , Lymph Nodes/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mutation , Receptors, Notch/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...