Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Diabetologia ; 66(10): 1925-1942, 2023 10.
Article in English | MEDLINE | ID: mdl-37480416

ABSTRACT

AIM/HYPOTHESIS: Hyperglycaemia is associated with alpha cell dysfunction, leading to dysregulated glucagon secretion in type 1 and type 2 diabetes; however, the mechanisms involved are still elusive. The nutrient sensor mammalian target of rapamycin complex 1 (mTORC1) plays a major role in the maintenance of alpha cell mass and function. We studied the regulation of alpha cell mTORC1 by nutrients and its role in the development of hyperglucagonaemia in diabetes. METHODS: Alpha cell mTORC1 activity was assessed by immunostaining for phosphorylation of its downstream target, the ribosomal protein S6, and glucagon, followed by confocal microscopy on pancreatic sections and flow cytometry on dispersed human and mouse islets and the alpha cell line, αTC1-6. Metabolomics and metabolic flux were studied by 13C glucose labelling in 2.8 or 16.7 mmol/l glucose followed by LC-MS analysis. To study the role of mTORC1 in mediating hyperglucagonaemia in diabetes, we generated an inducible alpha cell-specific Rptor knockout in the Akita mouse model of diabetes and tested the effects on glucose tolerance by IPGTT and on glucagon secretion. RESULTS: mTORC1 activity was increased in alpha cells from diabetic Akita mice in parallel to the development of hyperglycaemia and hyperglucagonaemia (two- to eightfold increase). Acute exposure of mouse and human islets to amino acids stimulated alpha cell mTORC1 (3.5-fold increase), whereas high glucose concentrations inhibited mTORC1 (1.4-fold decrease). The mTORC1 response to glucose was abolished in human and mouse diabetic alpha cells following prolonged islet exposure to high glucose levels, resulting in sustained activation of mTORC1, along with increased glucagon secretion. Metabolomics and metabolic flux analysis showed that exposure to high glucose levels enhanced glycolysis, glucose oxidation and the synthesis of glucose-derived amino acids. In addition, chronic exposure to high glucose levels increased the expression of Slc7a2 and Slc38a4, which encode amino acid transporters, as well as the levels of branched-chain amino acids and methionine cycle metabolites (~1.3-fold increase for both). Finally, conditional Rptor knockout in alpha cells from adult diabetic mice inhibited mTORC1, thereby inhibiting glucagon secretion (~sixfold decrease) and improving diabetes, despite persistent insulin deficiency. CONCLUSIONS/INTERPRETATION: Alpha cell exposure to hyperglycaemia enhances amino acid synthesis and transport, resulting in sustained activation of mTORC1, thereby increasing glucagon secretion. mTORC1 therefore plays a major role in mediating alpha cell dysfunction in diabetes. DATA AVAILABILITY: All sequencing data are available from the Gene Expression Omnibus (GEO) repository (accession no. GSE154126; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE154126 ).


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Hyperglycemia , Adult , Humans , Animals , Glucagon , Mechanistic Target of Rapamycin Complex 1 , Glucose , Mammals
2.
Cancer Res ; 82(22): 4164-4178, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36084256

ABSTRACT

Exercise prevents cancer incidence and recurrence, yet the underlying mechanism behind this relationship remains mostly unknown. Here we report that exercise induces the metabolic reprogramming of internal organs that increases nutrient demand and protects against metastatic colonization by limiting nutrient availability to the tumor, generating an exercise-induced metabolic shield. Proteomic and ex vivo metabolic capacity analyses of murine internal organs revealed that exercise induces catabolic processes, glucose uptake, mitochondrial activity, and GLUT expression. Proteomic analysis of routinely active human subject plasma demonstrated increased carbohydrate utilization following exercise. Epidemiologic data from a 20-year prospective study of a large human cohort of initially cancer-free participants revealed that exercise prior to cancer initiation had a modest impact on cancer incidence in low metastatic stages but significantly reduced the likelihood of highly metastatic cancer. In three models of melanoma in mice, exercise prior to cancer injection significantly protected against metastases in distant organs. The protective effects of exercise were dependent on mTOR activity, and inhibition of the mTOR pathway with rapamycin treatment ex vivo reversed the exercise-induced metabolic shield. Under limited glucose conditions, active stroma consumed significantly more glucose at the expense of the tumor. Collectively, these data suggest a clash between the metabolic plasticity of cancer and exercise-induced metabolic reprogramming of the stroma, raising an opportunity to block metastasis by challenging the metabolic needs of the tumor. SIGNIFICANCE: Exercise protects against cancer progression and metastasis by inducing a high nutrient demand in internal organs, indicating that reducing nutrient availability to tumor cells represents a potential strategy to prevent metastasis. See related commentary by Zerhouni and Piskounova, p. 4124.


Subject(s)
Exercise , Melanoma , Nutrients , Proteomics , Animals , Humans , Mice , Glucose/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Prospective Studies , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Exercise/physiology , Nutrients/genetics , Nutrients/metabolism
3.
Science ; 377(6601): 47-56, 2022 07.
Article in English | MEDLINE | ID: mdl-35771919

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) kinase controls growth in response to nutrients, including the amino acid leucine. In cultured cells, mTORC1 senses leucine through the leucine-binding Sestrin proteins, but the physiological functions and distribution of Sestrin-mediated leucine sensing in mammals are unknown. We find that mice lacking Sestrin1 and Sestrin2 cannot inhibit mTORC1 upon dietary leucine deprivation and suffer a rapid loss of white adipose tissue (WAT) and muscle. The WAT loss is driven by aberrant mTORC1 activity and fibroblast growth factor 21 (FGF21) production in the liver. Sestrin expression in the liver lobule is zonated, accounting for zone-specific regulation of mTORC1 activity and FGF21 induction by leucine. These results establish the mammalian Sestrins as physiological leucine sensors and reveal a spatial organization to nutrient sensing by the mTORC1 pathway.


Subject(s)
Diet , Leucine , Liver , Mechanistic Target of Rapamycin Complex 1 , Sestrins , Adipose Tissue, White/enzymology , Animals , Leucine/metabolism , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Sestrins/metabolism , Signal Transduction
4.
Article in English | MEDLINE | ID: mdl-32122884

ABSTRACT

Treatment of type 1 diabetes with insulin injection is expensive, complicated, and insufficient. While cadaveric islet transplantations coupled with immunosuppressants can cure diabetes, the scarcity of acceptable islets is problematic. Developmental research on pancreas formation has informed in vitro differentiation of human pluripotent stem cells into functional islets. Although generating ß cells from stem cells offers a potential cure for type 1 diabetes, several challenges remain, including protecting the cells from the immune system.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Insulin-Secreting Cells/cytology , Islets of Langerhans Transplantation/methods , Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation , Humans , Immune System , Immunosuppression Therapy , Insulin/metabolism , Mice , Pancreas , Risk , Stem Cell Transplantation
5.
Cell Rep ; 31(6): 107623, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32402282

ABSTRACT

Stem cell-derived ß (SC-ß) cells could provide unlimited human ß cells toward a curative diabetes treatment. Differentiation of SC-ß cells yields transplantable islets that secrete insulin in response to glucose challenges. Following transplantation into mice, SC-ß cell function is comparable to human islets, but the magnitude and consistency of response in vitro are less robust than observed in cadaveric islets. Here, we profile metabolism of SC-ß cells and islets to quantify their capacity to sense glucose and identify reduced anaplerotic cycling in the mitochondria as the cause of reduced glucose-stimulated insulin secretion in SC-ß cells. This activity can be rescued by challenging SC-ß cells with intermediate metabolites from the TCA cycle and late but not early glycolysis, downstream of the enzymes glyceraldehyde 3-phosphate dehydrogenase and phosphoglycerate kinase. Bypassing this metabolic bottleneck results in a robust, bi-phasic insulin release in vitro that is identical in magnitude to functionally mature human islets.


Subject(s)
B-Lymphocytes/metabolism , Glucose/metabolism , Glycolysis/genetics , Stem Cells/metabolism , Animals , Cell Differentiation , Humans , Mice
6.
Cell Metab ; 31(5): 1004-1016.e5, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32375022

ABSTRACT

A drastic transition at birth, from constant maternal nutrient supply in utero to intermittent postnatal feeding, requires changes in the metabolic system of the neonate. Despite their central role in metabolic homeostasis, little is known about how pancreatic ß cells adjust to the new nutritional challenge. Here, we find that after birth ß cell function shifts from amino acid- to glucose-stimulated insulin secretion in correlation with the change in the nutritional environment. This adaptation is mediated by a transition in nutrient sensitivity of the mTORC1 pathway, which leads to intermittent mTORC1 activity. Disrupting nutrient sensitivity of mTORC1 in mature ß cells reverts insulin secretion to a functionally immature state. Finally, manipulating nutrient sensitivity of mTORC1 in stem cell-derived ß cells in vitro strongly enhances their glucose-responsive insulin secretion. These results reveal a mechanism by which nutrients regulate ß cell function, thereby enabling a metabolic adaptation for the newborn.


Subject(s)
Glucose/metabolism , Nutrients/metabolism , Animals , Cells, Cultured , Humans , Insulin Secretion , Mice , Mice, Inbred C57BL
7.
Cell Stem Cell ; 26(1): 108-122.e10, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31839570

ABSTRACT

Stem-cell-derived tissues could transform disease research and therapy, yet most methods generate functionally immature products. We investigate how human pluripotent stem cells (hPSCs) differentiate into pancreatic islets in vitro by profiling DNA methylation, chromatin accessibility, and histone modification changes. We find that enhancer potential is reset upon lineage commitment and show how pervasive epigenetic priming steers endocrine cell fates. Modeling islet differentiation and maturation regulatory circuits reveals genes critical for generating endocrine cells and identifies circadian control as limiting for in vitro islet function. Entrainment to circadian feeding/fasting cycles triggers islet metabolic maturation by inducing cyclic synthesis of energy metabolism and insulin secretion effectors, including antiphasic insulin and glucagon pulses. Following entrainment, hPSC-derived islets gain persistent chromatin changes and rhythmic insulin responses with a raised glucose threshold, a hallmark of functional maturity, and function within days of transplantation. Thus, hPSC-derived tissues are amenable to functional improvement by circadian modulation.


Subject(s)
Cell Differentiation , Circadian Rhythm , Islets of Langerhans/cytology , Pluripotent Stem Cells/cytology , Glucagon/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism
8.
Circulation ; 141(4): 285-300, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31707831

ABSTRACT

BACKGROUND: Current differentiation protocols to produce cardiomyocytes from human induced pluripotent stem cells (iPSCs) are capable of generating highly pure cardiomyocyte populations as determined by expression of cardiac troponin T. However, these cardiomyocytes remain immature, more closely resembling the fetal state, with a lower maximum contractile force, slower upstroke velocity, and immature mitochondrial function compared with adult cardiomyocytes. Immaturity of iPSC-derived cardiomyocytes may be a significant barrier to clinical translation of cardiomyocyte cell therapies for heart disease. During development, cardiomyocytes undergo a shift from a proliferative state in the fetus to a more mature but quiescent state after birth. The mechanistic target of rapamycin (mTOR)-signaling pathway plays a key role in nutrient sensing and growth. We hypothesized that transient inhibition of the mTOR-signaling pathway could lead cardiomyocytes to a quiescent state and enhance cardiomyocyte maturation. METHODS: Cardiomyocytes were differentiated from 3 human iPSC lines using small molecules to modulate the Wnt pathway. Torin1 (0 to 200 nmol/L) was used to inhibit the mTOR pathway at various time points. We quantified contractile, metabolic, and electrophysiological properties of matured iPSC-derived cardiomyocytes. We utilized the small molecule inhibitor, pifithrin-α, to inhibit p53 signaling, and nutlin-3a, a small molecule inhibitor of MDM2 (mouse double minute 2 homolog) to upregulate and increase activation of p53. RESULTS: Torin1 (200 nmol/L) increased the percentage of quiescent cells (G0 phase) from 24% to 48% compared with vehicle control (P<0.05). Torin1 significantly increased expression of selected sarcomere proteins (including TNNI3 [troponin I, cardiac muscle]) and ion channels (including Kir2.1) in a dose-dependent manner when Torin1 was initiated after onset of cardiomyocyte beating. Torin1-treated cells had an increased relative maximum force of contraction, increased maximum oxygen consumption rate, decreased peak rise time, and increased downstroke velocity. Torin1 treatment increased protein expression of p53, and these effects were inhibited by pifithrin-α. In contrast, nutlin-3a independently upregulated p53, led to an increase in TNNI3 expression and worked synergistically with Torin1 to further increase expression of both p53 and TNNI3. CONCLUSIONS: Transient treatment of human iPSC-derived cardiomyocytes with Torin1 shifts cells to a quiescent state and enhances cardiomyocyte maturity.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Naphthyridines/pharmacology , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway/drug effects , Benzothiazoles/pharmacology , Cell Line , Humans , Imidazoles/pharmacology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Piperazines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors
9.
Curr Protoc Stem Cell Biol ; 51(1): e99, 2019 12.
Article in English | MEDLINE | ID: mdl-31756031

ABSTRACT

Our laboratory and others have developed protocols to generate glucose-responsive stem cell-derived ß cells in vitro. The cells resulting from these protocols could supplement or replace the use of human cadaveric islets for cell-based therapy for diabetes. The combination of an unlimited supply of pluripotent stem cell-derived ß cells and gene-editing approaches will facilitate numerous in vitro studies not possible with cadaveric islets. Here, we describe a protocol for fluorescent labeling and isolation of stem cell-derived ß cells. This purification of SC-ß cells is based on intracellular zinc content and is a simple method to complement other approaches for generating and assaying these cells. © 2019 The Authors. Basic Protocol: Fluorescent labeling and isolation of stem cell-derived ß cells.


Subject(s)
Insulin-Secreting Cells/cytology , Staining and Labeling/methods , Zinc/metabolism , Cell Culture Techniques/methods , Humans
10.
Dev Cell ; 45(6): 726-737.e3, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29920277

ABSTRACT

Developmental processes in different mammals are thought to share fundamental cellular mechanisms. We report a dramatic increase in cell size during postnatal pancreas development in rodents, accounting for much of the increase in organ size after birth. Hypertrophy of pancreatic acinar cells involves both higher ploidy and increased biosynthesis per genome copy; is maximal adjacent to islets, suggesting endocrine to exocrine communication; and is partly driven by weaning-related processes. In contrast to the situation in rodents, pancreas cell size in humans remains stable postnatally, indicating organ growth by pure hyperplasia. Pancreatic acinar cell volume varies 9-fold among 24 mammalian species analyzed, and shows a striking inverse correlation with organismal lifespan. We hypothesize that cellular hypertrophy is a strategy for rapid postnatal tissue growth, entailing life-long detrimental effects.


Subject(s)
Organ Size/physiology , Pancreas/growth & development , Pancreas/metabolism , Acinar Cells/physiology , Animals , Cell Enlargement , Cell Size , Humans , Hypertrophy , Insulin-Secreting Cells/physiology , Mice , Pancreas, Exocrine/physiology
11.
PLoS Genet ; 13(6): e1006860, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28628612

ABSTRACT

Dorsal closure (DC) is a developmental process in which two contralateral epithelial sheets migrate to seal a large hole in the dorsal ectoderm of the Drosophila embryo. Two signaling pathways act sequentially to orchestrate this dynamic morphogenetic process. First, c-Jun N-terminal kinase (JNK) signaling activity in the dorsal-most leading edge (LE) cells of the epidermis induces expression of decapentaplegic (dpp). Second, Dpp, a secreted TGF-ß homolog, triggers cell shape changes in the adjacent, ventrally located lateral epidermis, that guide the morphogenetic movements and cell migration mandatory for DC. Here we uncover a cell non-autonomous requirement for the Epidermal growth factor receptor (Egfr) pathway in the lateral epidermis for sustained dpp expression in the LE. Specifically, we demonstrate that Egfr pathway activity in the lateral epidermis prevents expression of the gene scarface (scaf), encoding a secreted antagonist of JNK signaling. In embryos with compromised Egfr signaling, upregulated Scaf causes reduction of JNK activity in LE cells, thereby impeding completion of DC. Our results identify a new developmental role for Egfr signaling in regulating epithelial plasticity via crosstalk with the JNK pathway.


Subject(s)
Drosophila Proteins/genetics , Embryonic Development/genetics , ErbB Receptors/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Receptors, Invertebrate Peptide/genetics , Serine Proteases/genetics , Animals , Drosophila Proteins/biosynthesis , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ectoderm/growth & development , Ectoderm/metabolism , Embryo, Nonmammalian , Epidermis/growth & development , Epidermis/metabolism , ErbB Receptors/biosynthesis , Gene Expression Regulation, Developmental , JNK Mitogen-Activated Protein Kinases/biosynthesis , Morphogenesis/genetics , Receptors, Invertebrate Peptide/biosynthesis , Serine Proteases/biosynthesis , Signal Transduction
12.
Diabetes ; 65(7): 2081-93, 2016 07.
Article in English | MEDLINE | ID: mdl-26993067

ABSTRACT

The molecular program underlying infrequent replication of pancreatic ß-cells remains largely inaccessible. Using transgenic mice expressing green fluorescent protein in cycling cells, we sorted live, replicating ß-cells and determined their transcriptome. Replicating ß-cells upregulate hundreds of proliferation-related genes, along with many novel putative cell cycle components. Strikingly, genes involved in ß-cell functions, namely, glucose sensing and insulin secretion, were repressed. Further studies using single-molecule RNA in situ hybridization revealed that in fact, replicating ß-cells double the amount of RNA for most genes, but this upregulation excludes genes involved in ß-cell function. These data suggest that the quiescence-proliferation transition involves global amplification of gene expression, except for a subset of tissue-specific genes, which are "left behind" and whose relative mRNA amount decreases. Our work provides a unique resource for the study of replicating ß-cells in vivo.


Subject(s)
Cell Division/genetics , Cell Proliferation/genetics , Insulin-Secreting Cells/metabolism , Transcriptome , Animals , Flow Cytometry , Gene Expression Regulation , Insulin-Secreting Cells/cytology , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Transgenic
13.
Nat Med ; 22(4): 412-20, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26950362

ABSTRACT

Cellular senescence is thought to contribute to age-associated deterioration of tissue physiology. The senescence effector p16(Ink4a) is expressed in pancreatic beta cells during aging and limits their proliferative potential; however, its effects on beta cell function are poorly characterized. We found that beta cell-specific activation of p16(Ink4a) in transgenic mice enhances glucose-stimulated insulin secretion (GSIS). In mice with diabetes, this leads to improved glucose homeostasis, providing an unexpected functional benefit. Expression of p16(Ink4a) in beta cells induces hallmarks of senescence--including cell enlargement, and greater glucose uptake and mitochondrial activity--which promote increased insulin secretion. GSIS increases during the normal aging of mice and is driven by elevated p16(Ink4a) activity. We found that islets from human adults contain p16(Ink4a)-expressing senescent beta cells and that senescence induced by p16(Ink4a) in a human beta cell line increases insulin secretion in a manner dependent, in part, on the activity of the mechanistic target of rapamycin (mTOR) and the peroxisome proliferator-activated receptor (PPAR)-γ proteins. Our findings reveal a novel role for p16(Ink4a) and cellular senescence in promoting insulin secretion by beta cells and in regulating normal functional tissue maturation with age.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Aging/pathology , Animals , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Glucose/metabolism , Humans , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/pathology , Mice , Mice, Transgenic , PPAR gamma/genetics , TOR Serine-Threonine Kinases/genetics
14.
Cancer Res ; 73(9): 2829-39, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23423975

ABSTRACT

Cellular senescence, a state of cell-cycle arrest accompanied by dramatic morphologic and metabolic changes, is a central means by which cells respond to physiologic stress and oncogene activity. Senescence is thought to play important roles in aging and in tumor suppression, yet the dynamics by which senescent cells are formed, their effects on tissue function and their eventual fate are poorly understood. To study cellular senescence within an adult tissue, we developed transgenic mice inducibly expressing p14(ARF) (human ortholog of murine p19(ARF)), a central activator of senescence. Induction of p14(ARF) in the epidermis rapidly led to widespread apoptosis and cell-cycle arrest, a stage that was transient, and was followed by p53-dependent cellular senescence. The endogenous Cdkn2a products p19(ARF) and p16(Ink4a) were activated by the transgenic p14(ARF) through p53, revealing a senescence-promoting feed-forward loop. Commitment of cells to senescence required continued p14(ARF) expression, indicating that entry into this state depends on a persistent signal. However, once formed, senescent cells were retained in the epidermis, often for weeks after transgene silencing, indicating an absence of an efficient rapidly acting mechanism for their removal. Stem cells in the hair follicle bulge were largely protected from apoptosis upon p14(ARF) induction, but irreversibly lost their ability to proliferate and initiate follicle growth. Interestingly, induction of epidermal hyperplasia prevented the appearance of senescent cells upon p14(ARF) induction. Our findings provide basic insights into the dynamics of cellular senescence, a central tumor- suppressive mechanism, and reveal the potential for prolonged retention of senescent cells within tissues.


Subject(s)
Apoptosis , Epidermis/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Animals , Cell Cycle , Cellular Senescence , Gene Silencing , Hair Follicle/metabolism , Humans , Mice , Mice, Transgenic , Models, Biological , RNA/metabolism , Transgenes , Tumor Suppressor Protein p14ARF/physiology , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/metabolism
15.
Development ; 139(16): 3032-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22791891

ABSTRACT

The dorsoventral (DV) axis of the Drosophila embryo is patterned by a nuclear gradient of the Rel family transcription factor, Dorsal (Dl), that activates or represses numerous target genes in a region-specific manner. Here, we demonstrate that signaling by receptor tyrosine kinases (RTK) reduces nuclear levels and transcriptional activity of Dl, both at the poles and in the mid-body of the embryo. These effects depend on wntD, which encodes a Dl antagonist belonging to the Wingless/Wnt family of secreted factors. Specifically, we show that, via relief of Groucho- and Capicua-mediated repression, the Torso and EGFR RTK pathways induce expression of WntD, which in turn limits Dl nuclear localization at the poles and along the DV axis. Furthermore, this RTK-dependent control of Dl is important for restricting expression of its targets in both contexts. Thus, our results reveal a new mechanism of crosstalk, whereby RTK signals modulate the spatial distribution and activity of a developmental morphogen in vivo.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/genetics , Body Patterning/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Feedback, Physiological , Gene Expression Regulation, Developmental , Genes, Insect , HMGB Proteins/genetics , HMGB Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Mutation , Nuclear Proteins/genetics , Phosphoproteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics
16.
Dev Biol ; 360(1): 208-15, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21983201

ABSTRACT

The Drosophila neuroectoderm is initially subdivided into three longitudinal domains that give rise to columns of neuroblasts. This subdivision is coordinately accomplished by the action of the signaling pathways, Dorsal and Epidermal Growth Factor Receptor (EGFR), in conjunction with the homeodomain proteins, Ventral nervous system defective, Intermediate neuroblasts defective (Ind) and Muscle Segment Homeobox. We previously demonstrated that Ind expression is activated in response to the EGFR pathway. Here we show that EGF signaling subsequently mediates the direct phosphorylation of Ind by MAP kinase, which enhances the capacity of Ind to repress target genes, such as achaete. Specifically, we show that reduced EGF signaling results in diminished repression of achaete in the intermediate column, despite the presence of high levels of Ind protein. We also demonstrate that ectopic activation of MAP kinase results in the lateral expansion of the Ind expression domain with a corresponding reduction in achaete expression. This regulation is also dependent on the co-repressor, Dichaete. Our data indicate that EGF signaling, acting through MAP kinase, impinges on multiple aspects of Ind regulatory activity. While it has been often demonstrated that MAP kinase phosphorylation of transcriptional repressors attenuates their repressor activity, here we provide an example of phosphorylation enhancing repressor activity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Homeodomain Proteins/metabolism , MAP Kinase Signaling System , Animals , Animals, Genetically Modified , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA Primers/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental , Genes, Insect , Homeodomain Proteins/genetics , Mutagenesis, Site-Directed , Neurogenesis , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism
17.
Curr Biol ; 21(13): 1102-10, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21683597

ABSTRACT

BACKGROUND: Signaling by receptor tyrosine kinase (RTK) pathways plays fundamental roles in processes of cell-fate determination, often through the induction of specific transcriptional responses. Yet it is not fully understood how continuous target gene expression, required for irreversible cell-fate specification, is preserved after RTK signaling has ended. Here we address this question using the Drosophila embryo, a model system that has been instrumental in elucidating the developmental functions of RTK signal transduction. RESULTS: The Groucho corepressor is phosphorylated and downregulated in response to RTK signaling. Here we show that RTK pathways use Groucho phosphorylation as a general mechanism for inducing expression of pathway target genes encoding cell-fate determinants as well as feedback antagonists, indicating that relief of Groucho-dependent repression is an integral element of RTK signaling networks. We further demonstrate that after mitogen-activated protein kinase (MAPK) has been deactivated, sustained phosphorylation of Groucho is essential for persistent RTK-induced target gene expression and cell-fate determination in several developmental contexts. CONCLUSIONS: Phosphorylation of Groucho by MAPK plays a dual role in the regulation of RTK responses: (1) it mediates rapid feedback inhibition, and (2) it provides a stable memory mechanism of past MAPK activity. We propose that, in this manner, phosphorylation of Groucho enables transiently active RTK pathways to fix the spatiotemporal expression profiles of downstream targets over time.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Drosophila/metabolism , Gene Expression Regulation, Developmental , Receptor Protein-Tyrosine Kinases/metabolism , Repressor Proteins/metabolism , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Drosophila/embryology , Drosophila/genetics , Feedback, Physiological , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology
18.
Development ; 138(5): 915-24, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21270056

ABSTRACT

RTK/Ras/MAPK signaling pathways play key functions in metazoan development, but how they control expression of downstream genes is not well understood. In Drosophila, it is generally assumed that most transcriptional responses to RTK signal activation depend on binding of Ets-family proteins to specific cis-acting sites in target enhancers. Here, we show that several Drosophila RTK pathways control expression of downstream genes through common octameric elements that are binding sites for the HMG-box factor Capicua, a transcriptional repressor that is downregulated by RTK signaling in different contexts. We show that Torso RTK-dependent regulation of terminal gap gene expression in the early embryo critically depends on Capicua octameric sites, and that binding of Capicua to these sites is essential for recruitment of the Groucho co-repressor to the huckebein enhancer in vivo. We then show that subsequent activation of the EGFR RTK pathway in the neuroectodermal region of the embryo controls dorsal-ventral gene expression by downregulating the Capicua protein, and that this control also depends on Capicua octameric motifs. Thus, a similar mechanism of RTK regulation operates during subdivision of the anterior-posterior and dorsal-ventral embryonic axes. We also find that identical DNA octamers mediate Capicua-dependent regulation of another EGFR target in the developing wing. Remarkably, a simple combination of activator-binding sites and Capicua motifs is sufficient to establish complex patterns of gene expression in response to both Torso and EGFR activation in different tissues. We conclude that Capicua octamers are general response elements for RTK signaling in Drosophila.


Subject(s)
DNA-Binding Proteins , Drosophila Proteins/genetics , HMGB Proteins/genetics , MAP Kinase Signaling System , Receptor Protein-Tyrosine Kinases/metabolism , Repressor Proteins/genetics , Response Elements , Animals , Binding Sites , Body Patterning , Drosophila , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental , Protein Multimerization , Wings, Animal/growth & development
19.
Methods Mol Biol ; 661: 401-8, 2010.
Article in English | MEDLINE | ID: mdl-20811997

ABSTRACT

In Drosophila, like in other metazoans, receptor tyrosine kinase (RTK) signaling pathways control diverse cellular processes such as migration, growth, fate determination, and differentiation (Shilo, Development 132:4017-4027, 2005). Activation of RTKs by their extracellular ligands triggers a signal transduction cascade, mediated by the Ras/Raf/MEK cassette, which ultimately leads to dual phosphorylation and activation of the mitogen-activated protein kinase/extracellularly regulated kinase (MAPK/Erk). Once active, MAPK/Erk phosphorylates its cytoplasmic and nuclear substrates, consequently modulating (i.e., stimulating or inhibiting) their biological function (Murphy and Blenis, Trends in Biochemical Sciences 31:268-275, 2006). The currently available antibody specific for the doubly phosphorylated form of MAPK/Erk (dpERK) (Yung et al., FEBS Letters 408:292-296, 1997) provides a valuable readout for RTK signaling: it enables the spatiotemporal detection of RTK pathway activity in the developing organism, in situ (Gabay et al., Development 124:3535-3541, 1997; Gabay et al., Science 277:1103-1106, 1997). Here, we present a detailed protocol for anti-dpERK immunofluorescent staining that can be applied to the analysis of MAPK/Erk signaling in Drosophila embryogenesis.


Subject(s)
Drosophila melanogaster/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique/methods , Phosphoproteins/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Staining and Labeling/methods , Animals , Antibodies/immunology , Antibodies/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/enzymology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Extracellular Signal-Regulated MAP Kinases/immunology , Tissue Fixation
20.
Development ; 135(5): 829-37, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18216172

ABSTRACT

RTK pathways establish cell fates in a wide range of developmental processes. However, how the pathway effector MAPK coordinately regulates the expression of multiple target genes is not fully understood. We have previously shown that the EGFR RTK pathway causes phosphorylation and downregulation of Groucho, a global co-repressor that is widely used by many developmentally important repressors for silencing their various targets. Here, we use specific antibodies that reveal the dynamics of Groucho phosphorylation by MAPK, and show that Groucho is phosphorylated in response to several RTK pathways during Drosophila embryogenesis. Focusing on the regulation of terminal patterning by the Torso RTK pathway, we demonstrate that attenuation of Groucho's repressor function via phosphorylation is essential for the transcriptional output of the pathway and for terminal cell specification. Importantly, Groucho is phosphorylated by an efficient mechanism that does not alter its subcellular localisation or decrease its stability; rather, modified Groucho endures long after MAPK activation has terminated. We propose that phosphorylation of Groucho provides a widespread, long-term mechanism by which RTK signals control target gene expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Drosophila/embryology , Gene Expression Regulation, Developmental , Receptor Protein-Tyrosine Kinases/metabolism , Repressor Proteins/genetics , Animals , Drosophila/enzymology , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/physiology , In Situ Hybridization , Kinetics , Larva/physiology , Neural Plate/physiology , Phosphorylation , Plasmids
SELECTION OF CITATIONS
SEARCH DETAIL
...