1
|
Xu X, Pots H, Gilsbach BK, Parsons D, Veltman DM, Ramachandra SG, Li H, Kortholt A, Jin T. C2GAP2 is a common regulator of Ras signaling for chemotaxis, phagocytosis, and macropinocytosis. Front Immunol 2022; 13:1075386. [PMID: 36524124 PMCID: PMC9745196 DOI: 10.3389/fimmu.2022.1075386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/16/2022] [Indexed: 11/30/2022] Open
Abstract
Phagocytosis, macropinocytosis, and G protein coupled receptor-mediated chemotaxis are Ras-regulated and actin-driven processes. The common regulator for Ras activity in these three processes remains unknown. Here, we show that C2GAP2, a Ras GTPase activating protein, highly expressed in the vegetative growth state in model organism Dictyostelium. C2GAP2 localizes at the leading edge of chemotaxing cells, phagosomes during phagocytosis, and macropinosomes during micropinocytosis. c2gapB- cells lacking C2GAP2 displayed increased Ras activation upon folic acid stimulation and subsequent impaired chemotaxis in the folic acid gradient. In addition, c2gaB- cells have elevated phagocytosis and macropinocytosis, which subsequently results in faster cell growth. C2GAP2 binds multiple phospholipids on the plasma membrane and the membrane recruitment of C2GAP2 requires calcium. Taken together, we show a shared negative regulator of Ras signaling that mediates Ras signaling for chemotaxis, phagocytosis, and macropinocytosis.
Collapse
Affiliation(s)
- Xuehua Xu
- Chemotaxis Signaling Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States,*Correspondence: Xuehua Xu,
| | - Henderikus Pots
- Department of Cell Biochemistry, University of Groningen, Groningen, Netherlands
| | - Bernd K. Gilsbach
- Functional Neuroproteomics and Translational Biomarkers in Neurodegenerative Diseases German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Dustin Parsons
- Chemotaxis Signaling Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Douwe M. Veltman
- Department of Cell Biochemistry, University of Groningen, Groningen, Netherlands
| | - Sharmila G. Ramachandra
- Chemotaxis Signaling Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Haoran Li
- Chemotaxis Signaling Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen, Groningen, Netherlands
| | - Tian Jin
- Chemotaxis Signaling Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
2
|
Dynamics of Actin Cytoskeleton and Their Signaling Pathways during Cellular Wound Repair. Cells 2022; 11:cells11193166. [PMID: 36231128 PMCID: PMC9564287 DOI: 10.3390/cells11193166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/17/2022] Open
Abstract
The repair of wounded cell membranes is essential for cell survival. Upon wounding, actin transiently accumulates at the wound site. The loss of actin accumulation leads to cell death. The mechanism by which actin accumulates at the wound site, the types of actin-related proteins participating in the actin remodeling, and their signaling pathways are unclear. We firstly examined how actin accumulates at a wound site in Dictyostelium cells. Actin assembled de novo at the wound site, independent of cortical flow. Next, we searched for actin- and signal-related proteins targeting the wound site. Fourteen of the examined proteins transiently accumulated at different times. Thirdly, we performed functional analyses using gene knockout mutants or specific inhibitors. Rac, WASP, formin, the Arp2/3 complex, profilin, and coronin contribute to the actin dynamics. Finally, we found that multiple signaling pathways related to TORC2, the Elmo/Doc complex, PIP2-derived products, PLA2, and calmodulin are involved in the actin dynamics for wound repair.
Collapse
|
3
|
Consalvo KM, Kirolos SA, Sestak CE, Gomer RH. Sex-Based Differences in Human Neutrophil Chemorepulsion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:354-367. [PMID: 35793910 PMCID: PMC9283293 DOI: 10.4049/jimmunol.2101103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/02/2022] [Indexed: 05/25/2023]
Abstract
A considerable amount is known about how eukaryotic cells move toward an attractant, and the mechanisms are conserved from Dictyostelium discoideum to human neutrophils. Relatively little is known about chemorepulsion, where cells move away from a repellent signal. We previously identified pathways mediating chemorepulsion in Dictyostelium, and here we show that these pathways, including Ras, Rac, protein kinase C, PTEN, and ERK1 and 2, are required for human neutrophil chemorepulsion, and, as with Dictyostelium chemorepulsion, PI3K and phospholipase C are not necessary, suggesting that eukaryotic chemorepulsion mechanisms are conserved. Surprisingly, there were differences between male and female neutrophils. Inhibition of Rho-associated kinases or Cdc42 caused male neutrophils to be more repelled by a chemorepellent and female neutrophils to be attracted to the chemorepellent. In the presence of a chemorepellent, compared with male neutrophils, female neutrophils showed a reduced percentage of repelled neutrophils, greater persistence of movement, more adhesion, less accumulation of PI(3,4,5)P3, and less polymerization of actin. Five proteins associated with chemorepulsion pathways are differentially abundant, with three of the five showing sex dimorphism in protein localization in unstimulated male and female neutrophils. Together, this indicates a fundamental difference in a motility mechanism in the innate immune system in men and women.
Collapse
Affiliation(s)
| | - Sara A Kirolos
- Department of Biology, Texas A&M University, College Station, TX
| | - Chelsea E Sestak
- Department of Biology, Texas A&M University, College Station, TX
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX
| |
Collapse
|
4
|
Kirolos SA, Gomer RH. A chemorepellent inhibits local Ras activation to inhibit pseudopod formation to bias cell movement away from the chemorepellent. Mol Biol Cell 2021; 33:ar9. [PMID: 34788129 PMCID: PMC8886819 DOI: 10.1091/mbc.e20-10-0656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The ability of cells to sense chemical gradients is essential during development, morphogenesis, and immune responses. Although much is known about chemoattraction, chemorepulsion remains poorly understood. Proliferating Dictyostelium cells secrete a chemorepellent protein called AprA. AprA prevents pseudopod formation at the region of the cell closest to the source of AprA, causing the random movement of cells to be biased away from the AprA. Activation of Ras proteins in a localized sector of a cell cortex helps to induce pseudopod formation, and Ras proteins are needed for AprA chemorepulsion. Here we show that AprA locally inhibits Ras cortical activation through the G protein–coupled receptor GrlH, the G protein subunits Gβ and Gα8, Ras protein RasG, protein kinase B, the p21-activated kinase PakD, and the extracellular signal–regulated kinase Erk1. Diffusion calculations and experiments indicate that in a colony of cells, high extracellular concentrations of AprA in the center can globally inhibit Ras activation, while a gradient of AprA that naturally forms at the edge of the colony allows cells to activate Ras at sectors of the cell other than the sector of the cell closest to the center of the colony, effectively inducing both repulsion from the colony and cell differentiation. Together, these results suggest that a pathway that inhibits local Ras activation can mediate chemorepulsion.
Collapse
Affiliation(s)
- Sara A Kirolos
- Department of Biology, Texas A&M University, 301 Old Main Drive, College Station, Texas, 77843-3474 USA
| | - Richard H Gomer
- Department of Biology, Texas A&M University, 301 Old Main Drive, College Station, Texas, 77843-3474 USA
| |
Collapse
|
5
|
SenGupta S, Parent CA, Bear JE. The principles of directed cell migration. Nat Rev Mol Cell Biol 2021; 22:529-547. [PMID: 33990789 PMCID: PMC8663916 DOI: 10.1038/s41580-021-00366-6] [Citation(s) in RCA: 271] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 02/03/2023]
Abstract
Cells have the ability to respond to various types of environmental cues, and in many cases these cues induce directed cell migration towards or away from these signals. How cells sense these cues and how they transmit that information to the cytoskeletal machinery governing cell translocation is one of the oldest and most challenging problems in biology. Chemotaxis, or migration towards diffusible chemical cues, has been studied for more than a century, but information is just now beginning to emerge about how cells respond to other cues, such as substrate-associated cues during haptotaxis (chemical cues on the surface), durotaxis (mechanical substrate compliance) and topotaxis (geometric features of substrate). Here we propose four common principles, or pillars, that underlie all forms of directed migration. First, a signal must be generated, a process that in physiological environments is much more nuanced than early studies suggested. Second, the signal must be sensed, sometimes by cell surface receptors, but also in ways that are not entirely clear, such as in the case of mechanical cues. Third, the signal has to be transmitted from the sensing modules to the machinery that executes the actual movement, a step that often requires amplification. Fourth, the signal has to be converted into the application of asymmetric force relative to the substrate, which involves mostly the cytoskeleton, but perhaps other players as well. Use of these four pillars has allowed us to compare some of the similarities between different types of directed migration, but also to highlight the remarkable diversity in the mechanisms that cells use to respond to different cues provided by their environment.
Collapse
Affiliation(s)
- Shuvasree SenGupta
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carole A Parent
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - James E Bear
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
6
|
An Autocrine Negative Feedback Loop Inhibits Dictyostelium discoideum Proliferation through Pathways Including IP3/Ca 2. mBio 2021; 12:e0134721. [PMID: 34154396 PMCID: PMC8262924 DOI: 10.1128/mbio.01347-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Little is known about how eukaryotic cells can sense their number or spatial density and stop proliferating when the local density reaches a set value. We previously found that Dictyostelium discoideum accumulates extracellular polyphosphate to inhibit its proliferation, and this requires the G protein-coupled receptor GrlD and the small GTPase RasC. Here, we show that cells lacking the G protein component Gβ, the Ras guanine nucleotide exchange factor GefA, phosphatase and tensin homolog (PTEN), phospholipase C (PLC), inositol 1,4,5-trisphosphate (IP3) receptor-like protein A (IplA), polyphosphate kinase 1 (Ppk1), or the TOR complex 2 component PiaA have significantly reduced sensitivity to polyphosphate-induced proliferation inhibition. Polyphosphate upregulates IP3, and this requires GrlD, GefA, PTEN, PLC, and PiaA. Polyphosphate also upregulates cytosolic Ca2+, and this requires GrlD, Gβ, GefA, RasC, PLC, IplA, Ppk1, and PiaA. Together, these data suggest that polyphosphate uses signal transduction pathways including IP3/Ca2+ to inhibit the proliferation of D. discoideum. IMPORTANCE Many mammalian tissues such as the liver have the remarkable ability to regulate their size and have their cells stop proliferating when the tissue reaches the correct size. One possible mechanism involves the cells secreting a signal that they all sense, and a high level of the signal tells the cells that there are enough of them and to stop proliferating. Although regulating such mechanisms could be useful to regulate tissue size to control cancer or birth defects, little is known about such systems. Here, we use a microbial system to study such a mechanism, and we find that key elements of the mechanism have similarities to human proteins. This then suggests the possibility that we may eventually be able to regulate the proliferation of selected cell types in humans and animals.
Collapse
|
7
|
Das R, Chinnathambi S. Microglial remodeling of actin network by Tau oligomers, via G protein-coupled purinergic receptor, P2Y12R-driven chemotaxis. Traffic 2021; 22:153-170. [PMID: 33527700 DOI: 10.1111/tra.12784] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/05/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is associated with age-related neurodegeneration, synaptic deformation and chronic inflammation mediated by microglia and infiltrated macrophages in the brain. Tau oligomers can be released from damaged neurons via various mechanisms such as exosomes, neurotransmitter, membrane leakage etc. Microglia sense the extracellular Tau through several cell-surface receptors and mediate chemotaxis and phagocytosis. The purinergic receptor P2Y12R recently gained interest in neurodegeneration for neuro-glial communication and microglial chemotaxis towards the site of plaque deposition. To understand the effect of extracellular Tau oligomers in microglial migration, the P2Y12R-mediated actin remodeling, reorientation of tubulin network and rate of migration were studied in the presence of ATP. The extracellular Tau species directly interacted with P2Y12R and also induced this purinoceptor expression in microglia. Microglial P2Y12R colocalized with remodeled membrane-associated actin network as a component of migration in response to Tau oligomers. As an inducer of P2Y12R, ATP facilitated the localization of P2Y12R in lamellipodia and filopodia during accelerated microglial migration. The direct interaction of extracellular Tau oligomers with microglial P2Y12R would facilitate the signal transduction in both way, directional chemotaxis and receptor-mediated phagocytosis. These unprecedented findings emphasize that microglia can modulate the membrane-associated actin structure and incorporate P2Y12R to perceive the axis and rate of chemotaxis in Tauopathy.
Collapse
Affiliation(s)
- Rashmi Das
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
8
|
van Haastert PJM. Unified control of amoeboid pseudopod extension in multiple organisms by branched F-actin in the front and parallel F-actin/myosin in the cortex. PLoS One 2020; 15:e0243442. [PMID: 33296414 PMCID: PMC7725310 DOI: 10.1371/journal.pone.0243442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
The trajectory of moving eukaryotic cells depends on the kinetics and direction of extending pseudopods. The direction of pseudopods has been well studied to unravel mechanisms for chemotaxis, wound healing and inflammation. However, the kinetics of pseudopod extension-when and why do pseudopods start and stop- is equally important, but is largely unknown. Here the START and STOP of about 4000 pseudopods was determined in four different species, at four conditions and in nine mutants (fast amoeboids Dictyostelium and neutrophils, slow mesenchymal stem cells, and fungus B.d. chytrid with pseudopod and a flagellum). The START of a first pseudopod is a random event with a probability that is species-specific (23%/s for neutrophils). In all species and conditions, the START of a second pseudopod is strongly inhibited by the extending first pseudopod, which depends on parallel filamentous actin/myosin in the cell cortex. Pseudopods extend at a constant rate by polymerization of branched F-actin at the pseudopod tip, which requires the Scar complex. The STOP of pseudopod extension is induced by multiple inhibitory processes that evolve during pseudopod extension and mainly depend on the increasing size of the pseudopod. Surprisingly, no differences in pseudopod kinetics are detectable between polarized, unpolarized or chemotactic cells, and also not between different species except for small differences in numerical values. This suggests that the analysis has uncovered the fundament of cell movement with distinct roles for stimulatory branched F-actin in the protrusion and inhibitory parallel F-actin in the contractile cortex.
Collapse
|
9
|
Das R, Chinnathambi S. Actin-mediated Microglial Chemotaxis via G-Protein Coupled Purinergic Receptor in Alzheimer's Disease. Neuroscience 2020; 448:325-336. [PMID: 32941933 DOI: 10.1016/j.neuroscience.2020.09.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease mainly associated with aging, oxidative stress and genetic mutations. There are two pathological proteins involved in AD; Amyloid-β peptide and microtubule-associated protein Tau (MAPT). The β- and γ-secretase enzyme cleaves the Amyloid precursor protein, which results in the formation of extracellular plaques in brain. While, Tau undergoes hyperphosphorylation and other post-translational modifications (PTMs), which eventually generates Tau oligomers, and intracellular neurofibrillary tangles (NFTs) in neurons. Moreover, the brain-resident glia and infiltrated macrophages elevate the level of CNS inflammation, which trigger the oxidative damage of neuronal circuits by reactive oxygen species (ROS) and Nitric oxide (NO). Microglia is the primary immune cell in the CNS, which is continuously surveilling the neuronal synapses and pathogen invasion. Microglia in the resting state is called 'Ramified', which possess long surveilling extensions with a small cell body. But, upon activation, microglia retracts the cellular extensions and transform into round migratory cells, called as 'Amoeboid' state. Activated microglia undergoes actin remodeling by forming lamellipodia and filopodia, which directs the migratory axis while podosomes formed are involved in extracellular matrix degradation for invasion. Protein-aggregates in malfunctioning synapses and in CNS milieu can be detected by microglia, which results in its activation and migration. Subsequently, the phagocytosis of synapses leads to the inflammatory burst and memory loss. The extracellular nucleotides released from damaged neurons and the cytokine-chemokine gradients allow the neighboring microglia and macrophages to migrate-infiltrate at the site of neuronal-damage. The ionotropic (P2XR) and metabotropic (P2YR) purinergic receptor recognize extracellular ATP/ADP, which propagates through the intracellular calcium signaling, chemotaxis, phagocytosis and inflammation. The P2Y receptors give 'find me' or 'eat me' signals to microglia to either migrate or phagocytose cellular debris. Further, the actin cytoskeleton helps microglia to mediate directed chemotaxis and neuronal repair during neurodegeneration. Hence, we aim to emphasize the connection between purinergic signaling and actin-driven mechanical movements of microglia for migration and inflammation in AD.
Collapse
Affiliation(s)
- Rashmi Das
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India.
| |
Collapse
|
10
|
Consalvo KM, Rijal R, Tang Y, Kirolos SA, Smith MR, Gomer RH. Extracellular signaling in Dictyostelium. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2020; 63:395-405. [PMID: 31840778 DOI: 10.1387/ijdb.190259rg] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the last few decades, we have learned a considerable amount about how eukaryotic cells communicate with each other, and what it is the cells are telling each other. The simplicity of Dictyostelium discoideum, and the wide variety of available tools to study this organism, makes it the equivalent of a hydrogen atom for cell and developmental biology. Studies using Dictyostelium have pioneered a good deal of our understanding of eukaryotic cell communication. In this review, we will present a brief overview of how Dictyostelium cells use extracellular signals to attract each other, repel each other, sense their local cell density, sense whether the nearby cells are starving or stressed, count themselves to organize the formation of structures containing a regulated number of cells, sense the volume they are in, and organize their multicellular development. Although we are probably just beginning to learn what the cells are telling each other, the elucidation of Dictyostelium extracellular signals has already led to the development of possible therapeutics for human diseases.
Collapse
Affiliation(s)
- Kristen M Consalvo
- Department of Biology, Texas A∧M University, College Station, Texas, USA
| | | | | | | | | | | |
Collapse
|
11
|
Chang FS, Wang Y, Dmitriev P, Gross J, Galione A, Pears C. A two-pore channel protein required for regulating mTORC1 activity on starvation. BMC Biol 2020; 18:8. [PMID: 31969153 PMCID: PMC6977259 DOI: 10.1186/s12915-019-0735-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/20/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Two-pore channels (TPCs) release Ca2+ from acidic intracellular stores and are implicated in a number of diseases, but their role in development is unclear. The social amoeba Dictyostelium discoideum proliferates as single cells that aggregate to form a multicellular organism on starvation. Starvation is sensed by the mTORC1 complex which, like TPC proteins, is found on acidic vesicles. Here, we address the role of TPCs in development and under starvation. RESULTS We report that disruption of the gene encoding the single Dictyostelium TPC protein, TPC2, leads to a delay in early development and prolonged growth in culture with delayed expression of early developmental genes, although a rapid starvation-induced increase in autophagy is still apparent. Ca2+ signals induced by extracellular cAMP are delayed in developing tpc2- cells, and aggregation shows increased sensitivity to weak bases, consistent with reduced acidity of the vesicles. In mammalian cells, the mTORC1 protein kinase has been proposed to suppress TPC channel opening. Here, we show a reciprocal effect as tpc2- cells show an increased level of phosphorylation of an mTORC1 substrate, 4E-BP1. mTORC1 inhibition reverses the prolonged growth and increases the efficiency of aggregation of tpc2- cells. CONCLUSION TPC2 is required for efficient growth development transition in Dictyostelium and acts through modulation of mTORC1 activity revealing a novel mode of regulation.
Collapse
Affiliation(s)
- Fu-Sheng Chang
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Yuntao Wang
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Phillip Dmitriev
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Julian Gross
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Catherine Pears
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
12
|
Hansen SD, Huang WYC, Lee YK, Bieling P, Christensen SM, Groves JT. Stochastic geometry sensing and polarization in a lipid kinase-phosphatase competitive reaction. Proc Natl Acad Sci U S A 2019; 116:15013-15022. [PMID: 31278151 PMCID: PMC6660746 DOI: 10.1073/pnas.1901744116] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Phosphorylation reactions, driven by competing kinases and phosphatases, are central elements of cellular signal transduction. We reconstituted a native eukaryotic lipid kinase-phosphatase reaction that drives the interconversion of phosphatidylinositol-4-phosphate [PI(4)P] and phosphatidylinositol-4,5-phosphate [PI(4,5)P2] on membrane surfaces. This system exhibited bistability and formed spatial composition patterns on supported membranes. In smaller confined regions of membrane, rapid diffusion ensures the system remains spatially homogeneous, but the final outcome-a predominantly PI(4)P or PI(4,5)P2 membrane composition-was governed by the size of the reaction environment. In larger confined regions, interplay between the reactions, diffusion, and confinement created a variety of differentially patterned states, including polarization. Experiments and kinetic modeling reveal how these geometric confinement effects arise from a mechanism based on stochastic fluctuations in the copy number of membrane-bound kinases and phosphatases. The underlying requirements for such behavior are unexpectedly simple and likely to occur in natural biological signaling systems.
Collapse
Affiliation(s)
- Scott D Hansen
- Department of Chemistry, University of California, Berkeley, CA 94720;
- California Institute for Quantitative Biosciences, Berkeley, CA 94720
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403
| | - William Y C Huang
- Department of Chemistry, University of California, Berkeley, CA 94720
| | - Young Kwang Lee
- Department of Chemistry, University of California, Berkeley, CA 94720
| | - Peter Bieling
- California Institute for Quantitative Biosciences, Berkeley, CA 94720
| | | | - Jay T Groves
- Department of Chemistry, University of California, Berkeley, CA 94720;
- California Institute for Quantitative Biosciences, Berkeley, CA 94720
| |
Collapse
|
13
|
Kolb AL, Hsu DW, Wallis ABA, Ura S, Rakhimova A, Pears CJ, Lakin ND. Dictyostelium as a Model to Assess Site-Specific ADP-Ribosylation Events. Methods Mol Biol 2019; 1813:125-148. [PMID: 30097865 DOI: 10.1007/978-1-4939-8588-3_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The amoeba Dictyostelium discoideum is a single-cell organism that can undergo a simple developmental program, making it an excellent model to study the molecular mechanisms of cell motility, signal transduction, and cell-type differentiation. A variety of human genes that are absent or show limited conservation in other invertebrate models have been identified in this organism. This includes ADP-ribosyltransferases, also known as poly-ADP-ribose polymerases (PARPs), a family of proteins that catalyze the addition of single or poly-ADP-ribose moieties onto target proteins. The genetic tractability of Dictyostelium and its relatively simple genome structure makes it possible to disrupt PARP gene combinations, in addition to specific ADP-ribosylation sites at endogenous loci. Together, this makes Dictyostelium an attractive model to assess how ADP-ribosylation regulates a variety of cellular processes including DNA repair, transcription, and cell-type specification. Here we describe a range of techniques to study ADP-ribosylation in Dictyostelium, including analysis of ADP-ribosylation events in vitro and in vivo, in addition to approaches to assess the functional roles of this modification in vivo.
Collapse
Affiliation(s)
- Anna-Lena Kolb
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Duen-Wei Hsu
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Ana B A Wallis
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Seiji Ura
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Alina Rakhimova
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
14
|
Fukushima S, Matsuoka S, Ueda M. Excitable dynamics of Ras triggers spontaneous symmetry breaking of PIP3 signaling in motile cells. J Cell Sci 2019; 132:jcs224121. [PMID: 30745337 PMCID: PMC6432713 DOI: 10.1242/jcs.224121] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/31/2019] [Indexed: 12/22/2022] Open
Abstract
Spontaneous cell movement is underpinned by an asymmetric distribution of signaling molecules including small G proteins and phosphoinositides on the cell membrane. However, the molecular network necessary for spontaneous symmetry breaking has not been fully elucidated. Here, we report that, in Dictyostelium discoideum, the spatiotemporal dynamics of GTP bound Ras (Ras-GTP) breaks the symmetry due its intrinsic excitability even in the absence of extracellular spatial cues and downstream signaling activities. A stochastic excitation of local and transient Ras activation induced phosphatidylinositol (3,4,5)-trisphosphate (PIP3) accumulation via direct interaction with Phosphoinositide 3-kinase (PI3K), causing tightly coupled traveling waves that propagated along the membrane. Comprehensive phase analysis of the waves of Ras-GTP and PIP3 metabolism-related molecules revealed the network structure of the excitable system including positive-feedback regulation of Ras-GTP by the downstream PIP3. A mathematical model reconstituted a series of the observed symmetry-breaking phenomena, illustrating the essential involvement of Ras excitability in the cellular decision-making process.
Collapse
Affiliation(s)
- Seiya Fukushima
- Department of Biological Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan
- RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka 565-0874, Japan
| | - Satomi Matsuoka
- RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka 565-0874, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahiro Ueda
- Department of Biological Science, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan
- RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka 565-0874, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
15
|
Rijal R, Consalvo KM, Lindsey CK, Gomer RH. An endogenous chemorepellent directs cell movement by inhibiting pseudopods at one side of cells. Mol Biol Cell 2018; 30:242-255. [PMID: 30462573 PMCID: PMC6589559 DOI: 10.1091/mbc.e18-09-0562] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Eukaryotic chemoattraction signal transduction pathways, such as those used by Dictyostelium discoideum to move toward cAMP, use a G protein-coupled receptor to activate multiple conserved pathways such as PI3 kinase/Akt/PKB to induce actin polymerization and pseudopod formation at the front of a cell, and PTEN to localize myosin II to the rear of a cell. Relatively little is known about chemorepulsion. We previously found that AprA is a chemorepellent protein secreted by Dictyostelium cells. Here we used 29 cell lines with disruptions of cAMP and/or AprA signal transduction pathway components, and delineated the AprA chemorepulsion pathway. We find that AprA uses a subset of chemoattraction signal transduction pathways including Ras, protein kinase A, target of rapamycin (TOR), phospholipase A, and ERK1, but does not require the PI3 kinase/Akt/PKB and guanylyl cyclase pathways to induce chemorepulsion. Possibly as a result of not using the PI3 kinase/Akt/PKB pathway and guanylyl cyclases, AprA does not induce actin polymerization or increase the pseudopod formation rate, but rather appears to inhibit pseudopod formation at the side of cells closest to the source of AprA.
Collapse
Affiliation(s)
- Ramesh Rijal
- Department of Biology, Texas A&M University, College Station, TX 77843-3474
| | - Kristen M Consalvo
- Department of Biology, Texas A&M University, College Station, TX 77843-3474
| | | | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843-3474
| |
Collapse
|
16
|
Matsuoka S, Ueda M. Mutual inhibition between PTEN and PIP3 generates bistability for polarity in motile cells. Nat Commun 2018; 9:4481. [PMID: 30367048 PMCID: PMC6203803 DOI: 10.1038/s41467-018-06856-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022] Open
Abstract
Phosphatidylinositol 3,4,5-trisphosphate (PIP3) and PIP3 phosphatase (PTEN) are enriched mutually exclusively on the anterior and posterior membranes of eukaryotic motile cells. However, the mechanism that causes this spatial separation between the two molecules is unknown. Here we develop a method to manipulate PIP3 levels in living cells and used it to show PIP3 suppresses the membrane localization of PTEN. Single-molecule measurements of membrane-association and -dissociation kinetics and of lateral diffusion reveal that PIP3 suppresses the PTEN binding site required for stable PTEN membrane binding. Mutual inhibition between PIP3 and PTEN provides a mechanistic basis for bistability that creates a PIP3-enriched/PTEN-excluded state and a PTEN-enriched/PIP3-excluded state underlying the strict spatial separation between PIP3 and PTEN. The PTEN binding site also mediates the suppression of PTEN membrane localization in chemotactic signaling. These results illustrate that the PIP3-PTEN bistable system underlies a cell's decision-making for directional movement irrespective of the environment.
Collapse
Affiliation(s)
- Satomi Matsuoka
- Laboratory for Cell Signaling Dynamics, RIKEN QBiC, 6-2-3, Furuedai, Suita, Osaka, 565-0874, Japan.
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Laboratory for Cell Signaling Dynamics, RIKEN BDR, 6-2-3, Furuedai, Suita, Osaka, 565-0874, Japan.
| | - Masahiro Ueda
- Laboratory for Cell Signaling Dynamics, RIKEN QBiC, 6-2-3, Furuedai, Suita, Osaka, 565-0874, Japan
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Laboratory of Single Molecule Biology, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
- Laboratory for Cell Signaling Dynamics, RIKEN BDR, 6-2-3, Furuedai, Suita, Osaka, 565-0874, Japan
| |
Collapse
|
17
|
Hickey K, Stabenfeldt SE. Using biomaterials to modulate chemotactic signaling for central nervous system repair. Biomed Mater 2018; 13:044106. [PMID: 29411713 PMCID: PMC5991092 DOI: 10.1088/1748-605x/aaad82] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chemotaxis enables cellular communication and movement within the body. This review focuses on exploiting chemotaxis as a tool for repair of the central nervous system (CNS) damaged from injury and/or degenerative diseases. Chemokines and factors alone may initiate repair following CNS injury/disease, but exogenous administration may enhance repair and promote regeneration. This review will discuss critical chemotactic molecules and factors that may promote neural regeneration. Additionally, this review highlights how biomaterials can impact the presentation and delivery of chemokines and growth factors to alter the regenerative response.
Collapse
Affiliation(s)
- Kassondra Hickey
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| | | |
Collapse
|
18
|
Mitochondrial Stress Tests Using Seahorse Respirometry on Intact Dictyostelium discoideum Cells. Methods Mol Biol 2017; 1407:41-61. [PMID: 27271893 DOI: 10.1007/978-1-4939-3480-5_4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mitochondria not only play a critical and central role in providing metabolic energy to the cell but are also integral to the other cellular processes such as modulation of various signaling pathways. These pathways affect many aspects of cell physiology, including cell movement, growth, division, differentiation, and death. Mitochondrial dysfunction which affects mitochondrial bioenergetics and causes oxidative phosphorylation defects can thus lead to altered cellular physiology and manifest in disease. The assessment of the mitochondrial bioenergetics can thus provide valuable insights into the physiological state, and the alterations to the state of the cells. Here, we describe a method to successfully use the Seahorse XF(e)24 Extracellular Flux Analyzer to assess the mitochondrial respirometry of the cellular slime mold Dictyostelium discoideum.
Collapse
|
19
|
Devreotes PN, Bhattacharya S, Edwards M, Iglesias PA, Lampert T, Miao Y. Excitable Signal Transduction Networks in Directed Cell Migration. Annu Rev Cell Dev Biol 2017; 33:103-125. [PMID: 28793794 DOI: 10.1146/annurev-cellbio-100616-060739] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Although directed migration of eukaryotic cells may have evolved to escape nutrient depletion, it has been adopted for an extensive range of physiological events during development and in the adult organism. The subversion of these movements results in disease, such as cancer. Mechanisms of propulsion and sensing are extremely diverse, but most eukaryotic cells move by extending actin-filled protrusions termed macropinosomes, pseudopodia, or lamellipodia or by extension of blebs. In addition to motility, directed migration involves polarity and directional sensing. The hundreds of gene products involved in these processes are organized into networks of parallel and interconnected pathways. Many of these components are activated or inhibited coordinately with stimulation and on each spontaneously extended protrusion. Moreover, these networks display hallmarks of excitability, including all-or-nothing responsiveness and wave propagation. Cellular protrusions result from signal transduction waves that propagate outwardly from an origin and drive cytoskeletal activity. The range of the propagating waves and hence the size of the protrusions can be altered by lowering or raising the threshold for network activation, with larger and wider protrusions favoring gliding or oscillatory behavior over amoeboid migration. Here, we evaluate the variety of models of excitable networks controlling directed migration and outline critical tests. We also discuss the utility of this emerging view in producing cell migration and in integrating the various extrinsic cues that direct migration.
Collapse
Affiliation(s)
- Peter N Devreotes
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205;
| | - Sayak Bhattacharya
- Department of Electrical and Computer Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218
| | - Marc Edwards
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205;
| | - Pablo A Iglesias
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205; .,Department of Electrical and Computer Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218
| | - Thomas Lampert
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205;
| | - Yuchuan Miao
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205;
| |
Collapse
|
20
|
Graziano BR, Gong D, Anderson KE, Pipathsouk A, Goldberg AR, Weiner OD. A module for Rac temporal signal integration revealed with optogenetics. J Cell Biol 2017; 216:2515-2531. [PMID: 28687663 PMCID: PMC5551696 DOI: 10.1083/jcb.201604113] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 11/23/2016] [Accepted: 05/18/2017] [Indexed: 01/07/2023] Open
Abstract
Dissecting the logic of individual signaling modules in complex networks can be challenging for cascades that exhibit feedback and redundancy. In this study, Graziano et al. take an optogenetics-based approach to identify and dissect a module that converts sustained PIP3 production to transient Rac activation in the neutrophil chemotaxis signaling network. Sensory systems use adaptation to measure changes in signaling inputs rather than absolute levels of signaling inputs. Adaptation enables eukaryotic cells to directionally migrate over a large dynamic range of chemoattractant. Because of complex feedback interactions and redundancy, it has been difficult to define the portion or portions of eukaryotic chemotactic signaling networks that generate adaptation and identify the regulators of this process. In this study, we use a combination of optogenetic intracellular inputs, CRISPR-based knockouts, and pharmacological perturbations to probe the basis of neutrophil adaptation. We find that persistent, optogenetically driven phosphatidylinositol (3,4,5)-trisphosphate (PIP3) production results in only transient activation of Rac, a hallmark feature of adaptive circuits. We further identify the guanine nucleotide exchange factor P-Rex1 as the primary PIP3-stimulated Rac activator, whereas actin polymerization and the GTPase-activating protein ArhGAP15 are essential for proper Rac turnoff. This circuit is masked by feedback and redundancy when chemoattractant is used as the input, highlighting the value of probing signaling networks at intermediate nodes to deconvolve complex signaling cascades.
Collapse
Affiliation(s)
- Brian R Graziano
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | - Delquin Gong
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | | | - Anne Pipathsouk
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | - Anna R Goldberg
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | - Orion D Weiner
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA .,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
21
|
Fan Y, Xie L, Chung CY. Signaling Pathways Controlling Microglia Chemotaxis. Mol Cells 2017; 40:163-168. [PMID: 28301917 PMCID: PMC5386953 DOI: 10.14348/molcells.2017.0011] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/05/2017] [Accepted: 03/07/2017] [Indexed: 12/20/2022] Open
Abstract
Microglia are the primary resident immune cells of the central nervous system (CNS). They are the first line of defense of the brain's innate immune response against infection, injury, and diseases. Microglia respond to extracellular signals and engulf unwanted neuronal debris by phagocytosis, thereby maintaining normal cellular homeostasis in the CNS. Pathological stimuli such as neuronal injury induce transformation and activation of resting microglia with ramified morphology into a motile amoeboid form and activated microglia chemotax toward lesion site. This review outlines the current research on microglial activation and chemotaxis.
Collapse
Affiliation(s)
- Yang Fan
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072,
P.R. China
| | - Lirui Xie
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072,
P.R. China
| | - Chang Y. Chung
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072,
P.R. China
| |
Collapse
|
22
|
Transcriptional profiling of rat skeletal muscle hypertrophy under restriction of blood flow. Gene 2016; 594:229-237. [PMID: 27613141 DOI: 10.1016/j.gene.2016.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 08/16/2016] [Accepted: 09/05/2016] [Indexed: 01/03/2023]
Abstract
Blood flow restriction (BFR) under low-intensity resistance training (LIRT) can produce similar effects upon muscles to that of high-intensity resistance training (HIRT) while overcoming many of the restrictions to HIRT that occurs in a clinical setting. However, the potential molecular mechanisms of BFR induced muscle hypertrophy remain largely unknown. Here, using a BFR rat model, we aim to better elucidate the mechanisms regulating muscle hypertrophy as induced by BFR and reveal possible clinical therapeutic targets for atrophy cases. We performed genome wide screening with microarray analysis to identify unique differentially expressed genes during rat muscle hypertrophy. We then successfully separated the differentially expressed genes from BRF treated soleus samples by comparing the Affymetrix rat Genome U34 2.0 array with the control. Using qRT-PCR and immunohistochemistry (IHC) we also analyzed other related differentially expressed genes. Results suggested that muscle hypertrophy induced by BFR is essentially regulated by the rate of protein turnover. Specifically, PI3K/AKT and MAPK pathways act as positive regulators in controlling protein synthesis where ubiquitin-proteasome acts as a negative regulator. This represents the first general genome wide level investigation of the gene expression profile in the rat soleus after BFR treatment. This may aid our understanding of the molecular mechanisms regulating and controlling muscle hypertrophy and provide support to the BFR strategies aiming to prevent muscle atrophy in a clinical setting.
Collapse
|
23
|
Lee SH, Sud N, Lee N, Subramaniyam S, Chung CY. Regulation of Integrin α6 Recycling by Calcium-independent Phospholipase A2 (iPLA2) to Promote Microglia Chemotaxis on Laminin. J Biol Chem 2016; 291:23645-23653. [PMID: 27655917 DOI: 10.1074/jbc.m116.732610] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Indexed: 11/06/2022] Open
Abstract
Microglia are the immune effector cells that are activated in response to pathological changes in the central nervous system. Microglial activation is accompanied by the alteration of integrin expression on the microglia surface. However, changes of integrin expression upon chemoattractant (ADP) stimulation still remain unknown. In this study, we investigated whether ADP induces the alteration of integrin species on the cell surface, leading to changes in chemotactic ability on different extracellular matrix proteins. Flow cytometry scans and on-cell Western assays showed that ADP stimulation induced a significant increase of α6 integrin-GFP, but not α5, on the surface of microglia cells. Microglia also showed a greater motility increase on laminin than fibronectin after ADP stimulation. Time lapse microscopy and integrin endocytosis assay revealed the essential role of calcium-independent phospholipase A2 activity for the recycling of α6 integrin-GFP from the endosomal recycling complex to the plasma membrane. Lack of calcium-independent phospholipase A2 activity caused a reduced rate of focal adhesion formation on laminin at the leading edge. Our results suggest that the alteration of integrin-mediated adhesion may regulate the extent of microglial infiltration into the site of damage by controlling their chemotactic ability.
Collapse
Affiliation(s)
- Sang-Hyun Lee
- From the Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600.,Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea, and
| | - Neetu Sud
- From the Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
| | - Narae Lee
- From the Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
| | - Selvaraj Subramaniyam
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Chang Y Chung
- From the Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600, .,School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
24
|
Pan M, Xu X, Chen Y, Jin T. Identification of a Chemoattractant G-Protein-Coupled Receptor for Folic Acid that Controls Both Chemotaxis and Phagocytosis. Dev Cell 2016; 36:428-39. [PMID: 26906738 DOI: 10.1016/j.devcel.2016.01.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/09/2015] [Accepted: 01/19/2016] [Indexed: 10/22/2022]
Abstract
Eukaryotic phagocytes search and destroy invading microorganisms via chemotaxis and phagocytosis. The social amoeba Dictyostelium discoideum is a professional phagocyte that chases bacteria through chemotaxis and engulfs them as food via phagocytosis. G-protein-coupled receptors (GPCRs) are known for detecting chemoattractants and directing cell migration, but their roles in phagocytosis are not clear. Here, we developed a quantitative phosphoproteomic technique to discover signaling components. Using this approach, we discovered the long sought after folic acid receptor, fAR1, in D. discoideum. We showed that the seven-transmembrane receptor fAR1 is required for folic acid-mediated signaling events. Significantly, we discovered that fAR1 is essential for both chemotaxis and phagocytosis of bacteria, thereby representing a chemoattractant GPCR that mediates not only chasing but also ingesting bacteria. We revealed that a phagocyte is able to internalize particles via a chemoattractant-mediated engulfment process. We propose that mammalian phagocytes may also use this mechanism to engulf and ingest bacterial pathogens.
Collapse
Affiliation(s)
- Miao Pan
- Chemotaxis Signal Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD 20852, USA.
| | - Xuehua Xu
- Chemotaxis Signal Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD 20852, USA
| | - Yong Chen
- Proteomics Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tian Jin
- Chemotaxis Signal Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Disease, NIH, Rockville, MD 20852, USA.
| |
Collapse
|
25
|
Takatori N, Oonuma K, Nishida H, Saiga H. Polarization of PI3K Activity Initiated by Ooplasmic Segregation Guides Nuclear Migration in the Mesendoderm. Dev Cell 2016; 35:333-43. [PMID: 26555053 DOI: 10.1016/j.devcel.2015.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/16/2015] [Accepted: 10/15/2015] [Indexed: 11/29/2022]
Abstract
Asymmetric localization of RNA is a widely observed mechanism of cell polarization. Using embryos of the ascidian, Halocynthia roretzi, we previously showed that mesoderm and endoderm fates are separated by localization of mRNA encoding a transcription factor, Not, to the future mesoderm-side cytoplasm of the mesendoderm cell through asymmetric positioning of the nucleus. Here, we investigated the mechanism that defines the direction of the nuclear migration. We show that localization of PtdIns(3,4,5)P3 to the future mesoderm region determines the direction of nuclear migration. Localization of PtdIns(3,4,5)P3 was dependent on the localization of PI3Kα to the future mesoderm region. PI3Kα was first localized at the 1-cell stage by the ooplasmic movement. Activity of localized PI3Kα at the 4-cell stage was required for the localization of PI3Kα up to the nuclear migration. Our results provide the scaffold for understanding the chain of causality leading to the separation of germ layer fates.
Collapse
Affiliation(s)
- Naohito Takatori
- Department of Biological Sciences, Graduate School of Science and Engineering, Tokyo Metropolitan University, 1-1 Minamiohsawa, Hachioji, Tokyo 192-0397, Japan; Department of Biological Sciences, Graduate school of Science, Osaka University 1-1, Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan.
| | - Kouhei Oonuma
- Department of Biological Sciences, Graduate School of Science and Engineering, Tokyo Metropolitan University, 1-1 Minamiohsawa, Hachioji, Tokyo 192-0397, Japan
| | - Hiroki Nishida
- Department of Biological Sciences, Graduate school of Science, Osaka University 1-1, Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Hidetoshi Saiga
- Department of Biological Sciences, Graduate School of Science and Engineering, Tokyo Metropolitan University, 1-1 Minamiohsawa, Hachioji, Tokyo 192-0397, Japan
| |
Collapse
|
26
|
Gβ Regulates Coupling between Actin Oscillators for Cell Polarity and Directional Migration. PLoS Biol 2016; 14:e1002381. [PMID: 26890004 PMCID: PMC4758609 DOI: 10.1371/journal.pbio.1002381] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/15/2016] [Indexed: 02/03/2023] Open
Abstract
For directional movement, eukaryotic cells depend on the proper organization of their actin cytoskeleton. This engine of motility is made up of highly dynamic nonequilibrium actin structures such as flashes, oscillations, and traveling waves. In Dictyostelium, oscillatory actin foci interact with signals such as Ras and phosphatidylinositol 3,4,5-trisphosphate (PIP3) to form protrusions. However, how signaling cues tame actin dynamics to produce a pseudopod and guide cellular motility is a critical open question in eukaryotic chemotaxis. Here, we demonstrate that the strength of coupling between individual actin oscillators controls cell polarization and directional movement. We implement an inducible sequestration system to inactivate the heterotrimeric G protein subunit Gβ and find that this acute perturbation triggers persistent, high-amplitude cortical oscillations of F-actin. Actin oscillators that are normally weakly coupled to one another in wild-type cells become strongly synchronized following acute inactivation of Gβ. This global coupling impairs sensing of internal cues during spontaneous polarization and sensing of external cues during directional motility. A simple mathematical model of coupled actin oscillators reveals the importance of appropriate coupling strength for chemotaxis: moderate coupling can increase sensitivity to noisy inputs. Taken together, our data suggest that Gβ regulates the strength of coupling between actin oscillators for efficient polarity and directional migration. As these observations are only possible following acute inhibition of Gβ and are masked by slow compensation in genetic knockouts, our work also shows that acute loss-of-function approaches can complement and extend the reach of classical genetics in Dictyostelium and likely other systems as well. Coupling of individual oscillators regulates biological functions ranging from crickets chirping in unison to the coordination of pacemaker cells of the heart. This study finds that a similar concept—coupling between actin oscillators—is at work within single slime mold cells to establish polarity and guide their direction of migration. The actin cytoskeleton of motile cells is comprised of highly dynamic structures. Recently, small oscillating actin foci have been discovered around the periphery of Dictyostelium cells. These oscillators are thought to enable pseudopod formation, but how their dynamics are regulated for this is unknown. Here, we demonstrate that the strength of coupling between individual actin oscillators controls cell polarization and directional movement. Actin oscillators are weakly coupled to one another in wild-type cells, but they become strongly synchronized after acute inactivation of the signaling protein Gβ. This global coupling impairs sensing of internal cues during spontaneous polarization and sensing of external cues during directional motility. Supported by a mathematical model, our data suggest that wild-type cells are tuned to an optimal coupling strength for patterning by upstream cues. These observations are only possible following acute inhibition of Gβ, which highlights the value of revisiting classical mutants with acute loss-of-function perturbations.
Collapse
|
27
|
Puniya BL, Allen L, Hochfelder C, Majumder M, Helikar T. Systems Perturbation Analysis of a Large-Scale Signal Transduction Model Reveals Potentially Influential Candidates for Cancer Therapeutics. Front Bioeng Biotechnol 2016; 4:10. [PMID: 26904540 PMCID: PMC4750464 DOI: 10.3389/fbioe.2016.00010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/25/2016] [Indexed: 12/20/2022] Open
Abstract
Dysregulation in signal transduction pathways can lead to a variety of complex disorders, including cancer. Computational approaches such as network analysis are important tools to understand system dynamics as well as to identify critical components that could be further explored as therapeutic targets. Here, we performed perturbation analysis of a large-scale signal transduction model in extracellular environments that stimulate cell death, growth, motility, and quiescence. Each of the model’s components was perturbed under both loss-of-function and gain-of-function mutations. Using 1,300 simulations under both types of perturbations across various extracellular conditions, we identified the most and least influential components based on the magnitude of their influence on the rest of the system. Based on the premise that the most influential components might serve as better drug targets, we characterized them for biological functions, housekeeping genes, essential genes, and druggable proteins. The most influential components under all environmental conditions were enriched with several biological processes. The inositol pathway was found as most influential under inactivating perturbations, whereas the kinase and small lung cancer pathways were identified as the most influential under activating perturbations. The most influential components were enriched with essential genes and druggable proteins. Moreover, known cancer drug targets were also classified in influential components based on the affected components in the network. Additionally, the systemic perturbation analysis of the model revealed a network motif of most influential components which affect each other. Furthermore, our analysis predicted novel combinations of cancer drug targets with various effects on other most influential components. We found that the combinatorial perturbation consisting of PI3K inactivation and overactivation of IP3R1 can lead to increased activity levels of apoptosis-related components and tumor-suppressor genes, suggesting that this combinatorial perturbation may lead to a better target for decreasing cell proliferation and inducing apoptosis. Finally, our approach shows a potential to identify and prioritize therapeutic targets through systemic perturbation analysis of large-scale computational models of signal transduction. Although some components of the presented computational results have been validated against independent gene expression data sets, more laboratory experiments are warranted to more comprehensively validate the presented results.
Collapse
Affiliation(s)
- Bhanwar Lal Puniya
- Department of Biochemistry, University of Nebraska-Lincoln , Lincoln, NE , USA
| | - Laura Allen
- Department of Mathematics, University of Nebraska at Omaha , Omaha, NE , USA
| | | | - Mahbubul Majumder
- Department of Mathematics, University of Nebraska at Omaha , Omaha, NE , USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln , Lincoln, NE , USA
| |
Collapse
|
28
|
Abstract
BACKGROUND The role of calcium-independent phospholipase A2 (iPLA2), a component of the three major PLA2 families, in acute/chronic inflammatory processes remains elusive. Previous investigations have documented iPLA2-mediated respiratory burst of neutrophils (PMNs); however, the causative isoform of iPLA2 is unidentified. We also demonstrated that the iPLA2γ-specific inhibitor attenuates trauma/hemorrhagic shock-induced lung injury. Therefore, iPLA2γ may be implicated in acute inflammation. In addition, arachidonic acid (AA), which is primarily produced by cytosolic PLA2 (cPLA2), is known to display PMN cytotoxicity, although the relationship between AA and the cytotoxic function is still being debated on. We therefore hypothesized that iPLA2γ regulates PMN cytotoxicity via AA-independent signaling pathways. The study aim was to distinguish the role of intracellular phospholipases A2, iPLA2, and cPLA2, in human PMN cytotoxicity and explore the possibility of the presence of signaling molecule(s) other than AA. METHODS Isolated human PMNs were incubated with the PLA2 inhibitor selective for iPLA2β, iPLA2γ, or cPLA2 and then activated with formyl-methionyl-leucyl-phenylalanine (fMLP) or phorbol 12-myristate 13-acetate (PMA). Superoxide production was assayed according to the superoxide dismutase-inhibitable cytochrome c reduction method, and the degree of elastase release was measured using a p-nitroanilide-conjugated elastase-specific substrate. In addition, chemotaxis toward platelet activating factor/fMLP was determined with a modified Boyden chamber system. RESULTS The iPLA2γ-specific inhibitor reduced the fMLP/PMA-stimulated superoxide generation by 90% and 30%, respectively; in addition, the inhibitor completely blocked the fMLP/PMA-activated elastase release. However, the cPLA2-specific inhibitor did not abrogate these effects to any degree at all concentrations. Likewise, the inhibitor for iPLA2γ, but not iPLA2β or cPLA2, completely inhibited the platelet activating factor/fMLP-induced chemotaxis. CONCLUSION iPLA2 is involved in extracellular reactive oxygen species production, elastase release, and chemotaxis in response to well-defined stimuli. In addition, the ineffectiveness of the cPLA2 inhibitor suggests that AA may not be relevant to these cytotoxic functions.
Collapse
|
29
|
Abstract
SUMMARY Stimuli that promote cell migration, such as chemokines, cytokines, and growth factors in metazoans and cyclic AMP in Dictyostelium, activate signaling pathways that control organization of the actin cytoskeleton and adhesion complexes. The Rho-family GTPases are a key convergence point of these pathways. Their effectors include actin regulators such as formins, members of the WASP/WAVE family and the Arp2/3 complex, and the myosin II motor protein. Pathways that link to the Rho GTPases include Ras GTPases, TorC2, and PI3K. Many of the molecules involved form gradients within cells, which define the front and rear of migrating cells, and are also established in related cellular behaviors such as neuronal growth cone extension and cytokinesis. The signaling molecules that regulate migration can be integrated to provide a model of network function. The network displays biochemical excitability seen as spontaneous waves of activation that propagate along the cell cortex. These events coordinate cell movement and can be biased by external cues to bring about directed migration.
Collapse
Affiliation(s)
- Peter Devreotes
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Alan Rick Horwitz
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| |
Collapse
|
30
|
Warren SC, Margineanu A, Katan M, Dunsby C, French PMW. Homo-FRET Based Biosensors and Their Application to Multiplexed Imaging of Signalling Events in Live Cells. Int J Mol Sci 2015; 16:14695-716. [PMID: 26133241 PMCID: PMC4519867 DOI: 10.3390/ijms160714695] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 12/20/2022] Open
Abstract
Multiplexed imaging of Förster Resonance Energy Transfer (FRET)-based biosensors potentially presents a powerful approach to monitoring the spatio-temporal correlation of signalling pathways within a single live cell. Here, we discuss the potential of homo-FRET based biosensors to facilitate multiplexed imaging. We demonstrate that the homo-FRET between pleckstrin homology domains of Akt (Akt-PH) labelled with mCherry may be used to monitor 3'-phosphoinositide accumulation in live cells and show how global analysis of time resolved fluorescence anisotropy measurements can be used to quantify this accumulation. We further present multiplexed imaging readouts of calcium concentration, using fluorescence lifetime measurements of TN-L15-a CFP/YFP based hetero-FRET calcium biosensor-with 3'-phosphoinositide accumulation.
Collapse
Affiliation(s)
- Sean C Warren
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK.
| | - Anca Margineanu
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK.
| | - Matilda Katan
- Structural and Molecular Biology, University College London, London WC1E 6BT, UK.
| | - Chris Dunsby
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK.
| | - Paul M W French
- Photonics Group, Department of Physics, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
31
|
Wu CY, Lin MW, Wu DC, Huang YB, Huang HT, Chen CL. The role of phosphoinositide-regulated actin reorganization in chemotaxis and cell migration. Br J Pharmacol 2014; 171:5541-54. [PMID: 25420930 DOI: 10.1111/bph.12777] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 04/15/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Reorganization of the actin cytoskeleton is essential for cell motility and chemotaxis. Actin-binding proteins (ABPs) and membrane lipids, especially phosphoinositides PI(4,5)P2 and PI(3,4,5)P3 are involved in the regulation of this reorganization. At least 15 ABPs have been reported to interact with, or regulated by phosphoinositides (PIPs) whose synthesis is regulated by extracellular signals. Recent studies have uncovered several parallel intracellular signalling pathways that crosstalk in chemotaxing cells. Here, we review the roles of ABPs and phosphoinositides in chemotaxis and cell migration. LINKED ARTICLES This article is part of a themed section on Cytoskeleton, Extracellular Matrix, Cell Migration, Wound Healing and Related Topics. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-24.
Collapse
Affiliation(s)
- C-Y Wu
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, Taiwan
| | | | | | | | | | | |
Collapse
|
32
|
Ren A, Moon C, Zhang W, Sinha C, Yarlagadda S, Arora K, Wang X, Yue J, Parthasarathi K, Heil-Chapdelaine R, Tigyi G, Naren AP. Asymmetrical macromolecular complex formation of lysophosphatidic acid receptor 2 (LPA2) mediates gradient sensing in fibroblasts. J Biol Chem 2014; 289:35757-69. [PMID: 25542932 DOI: 10.1074/jbc.m114.595512] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chemotactic migration of fibroblasts toward growth factors relies on their capacity to sense minute extracellular gradients and respond to spatially confined receptor-mediated signals. Currently, mechanisms underlying the gradient sensing of fibroblasts remain poorly understood. Using single-particle tracking methodology, we determined that a lysophosphatidic acid (LPA) gradient induces a spatiotemporally restricted decrease in the mobility of LPA receptor 2 (LPA2) on chemotactic fibroblasts. The onset of decreased LPA2 mobility correlates to the spatial recruitment and coupling to LPA2-interacting proteins that anchor the complex to the cytoskeleton. These localized PDZ motif-mediated macromolecular complexes of LPA2 trigger a Ca(2+) puff gradient that governs gradient sensing and directional migration in response to LPA. Disruption of the PDZ motif-mediated assembly of the macromolecular complex of LPA2 disorganizes the gradient of Ca(2+) puffs, disrupts gradient sensing, and reduces the directional migration of fibroblasts toward LPA. Our findings illustrate that the asymmetric macromolecular complex formation of chemoattractant receptors mediates gradient sensing and provides a new mechanistic basis for models to describe gradient sensing of fibroblasts.
Collapse
Affiliation(s)
- Aixia Ren
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Changsuk Moon
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Weiqiang Zhang
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Chandrima Sinha
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Sunitha Yarlagadda
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Kavisha Arora
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Xusheng Wang
- the Department of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, and
| | - Junming Yue
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Kaushik Parthasarathi
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | | | - Gabor Tigyi
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Anjaparavanda P Naren
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, the Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229,
| |
Collapse
|
33
|
Liu D, Yu Y, Schachner M. Ptena, but not Ptenb, reduces regeneration after spinal cord injury in adult zebrafish. Exp Neurol 2014; 261:196-205. [PMID: 24929056 DOI: 10.1016/j.expneurol.2014.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/04/2014] [Indexed: 02/05/2023]
Abstract
Based on the observation that the tumor suppressor gene PTEN (phosphatase and tensin homolog) reduces regeneration after spinal cord injury (SCI) as evidenced in the PTEN knockout mouse, we have investigated the function of Ptena and Ptenb, the two zebrafish homologs of mammalian PTEN, in adult zebrafish after spinal cord injury with the aim to assess the contribution of the two zebrafish genes to functional recovery in an animal species that spontaneously recovers from central nervous system injury. The inhibition of Ptena expression by antisense morpholino (MO) application improved spinal cord regeneration through 4 to 5weeks after injury. Retrograde tracing showed regrowth of axons from neurons of the regeneration-competent nucleus of the medial longitudinal fascicle in the brainstem in the Ptena MO-treated fish. Ptenb MO-treated fish recovered as well as control MO-treated fish at 4 and 5weeks after SCI, with their locomotion being similar to that of sham-injured and non-injured fish. The mRNA levels of Ptena were upregulated after SCI at the early stage after injury (12h and 6days) caudal to the lesion site, compared to the non-injured control, while the levels of Ptenb were upregulated only at 12h after injury. In situ hybridization experiments were in agreement with the qPCR measurements. At the protein level, Ptena was found to be expressed in spinal motoneurons and immature neurons. These results indicate that Ptena, but not Ptenb, inhibits regeneration in zebrafish, thus sharing this feature with PTEN in mammals. The fact that zebrafish regenerate better than mammals despite the inhibitory presence of Ptena is likely due to regeneration-conducive molecules that tip the balance from inhibition to enhancement. Interestingly, although Ptena and Ptenb have been shown to be functionally redundant in promoting the development of the fish larval central nervous system, they are not functionally redundant in the adult, suggesting that regeneration in fish is not predominantly due to the overall recapitulation of development.
Collapse
Affiliation(s)
- Dan Liu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong Province 515041, PR China
| | - Yong Yu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong Province 515041, PR China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong Province 515041, PR China.
| |
Collapse
|
34
|
Yan J, Jin T. Signaling network from GPCR to the actin cytoskeleton during chemotaxis. BIOARCHITECTURE 2014; 2:15-18. [PMID: 22754623 PMCID: PMC3383712 DOI: 10.4161/bioa.19740] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chemotaxis is crucial for many physiological processes including the recruitment of leukocytes to sites of infection, trafficking of lymphocytes in the human body, and metastasis of cancer cells. A family of small proteins, chemokines, serves as the signals, and a family of G-protein coupled receptors (GPCRs) detects chemokines and direct cell migration. One of the basic questions in chemotaxis of eukaryotes is how a GPCR transduces signals to control the assembly of the actin network that generates directional force for cell migration. Over the past decade, a variety of signaling components have been implicated to transduce the GPCR signaling to the actin cytoskeleton. Studies in a lower eukaryotic organism, Dictyostelium discoideum, have allowed us to discover evolutionary conversed components involved in the GPCR-controlled actin network during chemotaxis. However, complete pathways linking GPCR to the actin network are still far from clear. Here we first summarize the previous studies on these components, and then update with our finding showing a new pathway, consisting of a GPCR, Gβγ, Elmo/Dock, Rac and Arp2/3 and actin. We suggest that this pathway serves as a direct linkage between the GPCR/G-protein, the chemoattractant sensing machinery, and the actin cytoskeleton, the machinery of cell movement during chemotaxis of eukaryotic cells.
Collapse
Affiliation(s)
- Jianshe Yan
- Chemotaxis Signal Section; Laboratory of Immunogenetics; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Rockville, MD USA
| | | |
Collapse
|
35
|
Vorotnikov AV, Tyurin-Kuzmin PA. Chemotactic signaling in mesenchymal cells compared to amoeboid cells. Genes Dis 2014; 1:162-173. [PMID: 30258862 PMCID: PMC6150068 DOI: 10.1016/j.gendis.2014.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/15/2014] [Indexed: 01/09/2023] Open
Abstract
Cell chemotaxis plays a pivotal role in normal development, inflammatory response, injury repair and tissue regeneration in all organisms. It is also a critical contributor to cancer metastasis, altered angiogenesis and neurite growth in disease. The molecular mechanisms regulating chemotaxis are currently being identified and key components may be pertinent therapeutic targets. Although these components appear to be mostly common in various cells, there are important differences in chemotactic signaling networks and signal processing that result in the distinct chemotactic behavior of mesenchymal cells compared to much better studied amoeboid blood cells. These differences are not necessarily predetermined based on cell type, but are rather chosen and exploited by cells to modify their chemotactic behavior based on physical constraints and/or environmental conditions. This results in a specific type of chemotactic migration in mesenchymal cells that can be selectively targeted in disease. Here, we compare the chemotactic behavior, signaling and motility of mesenchymal and amoeboid cells. We suggest that the current model of chemotaxis is applicable for small amoeboid cells but needs to be reconsidered for large mesenchymal cells. We focus on new candidate regulatory molecules and feedback mechanisms that may account for mesenchymal cell type-specific chemotaxis.
Collapse
Key Words
- Chemotaxis
- Feedback regulation
- Fibroblasts
- GEFs, guanine nucleotide exchange factors
- GPCRs, G-protein coupled receptors
- Hydrogen peroxide
- LEGI, local excitation and global inhibition
- MAP-kinase, mitogen-activated protein kinase
- NOX, NADPH-oxidase
- PDGF, platelet derived growth factor
- PI3-kinase, phosphatidylinositol-3-kinase
- PIP3, phosphatidylinositol (3,4,5)-trisphosphate
- PLA2, phospholipase A2
- PTEN, phosphatase and tensin homolog
- RTKs, receptor tyrosine kinases
- Signaling
- mTORC, mechanistic target of rapamycin complex
- РТР-1В, protein tyrosine phosphatase-1B
Collapse
Affiliation(s)
- Alexander V. Vorotnikov
- Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University, Moscow, Russian Federation
- Institute of Experimental Cardiology, Russian Cardiology Research and Production Complex, Moscow, Russian Federation
- Corresponding author. Department of Biochemistry and Molecular Medicine, Faculty of Fundamental Medicine, Moscow State University, 31 Lomonosov Ave., Bldg 5, Russian Federation.
| | - Pyotr A. Tyurin-Kuzmin
- Faculty of Fundamental Medicine, M.V. Lomonosov Moscow State University, Moscow, Russian Federation
| |
Collapse
|
36
|
Hoeller O, Gong D, Weiner OD. How to understand and outwit adaptation. Dev Cell 2014; 28:607-616. [PMID: 24697896 DOI: 10.1016/j.devcel.2014.03.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 12/31/2022]
Abstract
Adaptation is the ability of a system to respond and reset itself even in the continuing presence of a stimulus. On one hand, adaptation is a physiological necessity that enables proper neuronal signaling and cell movement. On the other hand, adaptation can be a source of annoyance, as it can make biological systems resistant to experimental perturbations. Here we speculate where adaptation might live in eukaryotic chemotaxis and how it can be encoded in the signaling network. We then discuss tools and strategies that can be used to both understand and outwit adaptation in a wide range of cellular contexts.
Collapse
Affiliation(s)
- Oliver Hoeller
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158 USA
| | - Delquin Gong
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158 USA
| | - Orion D Weiner
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158 USA
| |
Collapse
|
37
|
Moving towards a paradigm: common mechanisms of chemotactic signaling in Dictyostelium and mammalian leukocytes. Cell Mol Life Sci 2014; 71:3711-47. [PMID: 24846395 DOI: 10.1007/s00018-014-1638-8] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/24/2014] [Accepted: 04/29/2014] [Indexed: 12/31/2022]
Abstract
Chemotaxis, or directed migration of cells along a chemical gradient, is a highly coordinated process that involves gradient sensing, motility, and polarity. Most of our understanding of chemotaxis comes from studies of cells undergoing amoeboid-type migration, in particular the social amoeba Dictyostelium discoideum and leukocytes. In these amoeboid cells the molecular events leading to directed migration can be conceptually divided into four interacting networks: receptor/G protein, signal transduction, cytoskeleton, and polarity. The signal transduction network occupies a central position in this scheme as it receives direct input from the receptor/G protein network, as well as feedback from the cytoskeletal and polarity networks. Multiple overlapping modules within the signal transduction network transmit the signals to the actin cytoskeleton network leading to biased pseudopod protrusion in the direction of the gradient. The overall architecture of the networks, as well as the individual signaling modules, is remarkably conserved between Dictyostelium and mammalian leukocytes, and the similarities and differences between the two systems are the subject of this review.
Collapse
|
38
|
Pears CJ, Lakin ND. Emerging models for DNA repair: Dictyostelium discoideum as a model for nonhomologous end-joining. DNA Repair (Amst) 2014; 17:121-31. [DOI: 10.1016/j.dnarep.2014.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 01/09/2014] [Accepted: 01/24/2014] [Indexed: 02/03/2023]
|
39
|
Zatulovskiy E, Tyson R, Bretschneider T, Kay RR. Bleb-driven chemotaxis of Dictyostelium cells. ACTA ACUST UNITED AC 2014; 204:1027-44. [PMID: 24616222 PMCID: PMC3998804 DOI: 10.1083/jcb.201306147] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Blebs and F-actin-driven pseudopods are alternative ways of extending the leading edge of migrating cells. We show that Dictyostelium cells switch from using predominantly pseudopods to blebs when migrating under agarose overlays of increasing stiffness. Blebs expand faster than pseudopods leaving behind F-actin scars, but are less persistent. Blebbing cells are strongly chemotactic to cyclic-AMP, producing nearly all of their blebs up-gradient. When cells re-orientate to a needle releasing cyclic-AMP, they stereotypically produce first microspikes, then blebs and pseudopods only later. Genetically, blebbing requires myosin-II and increases when actin polymerization or cortical function is impaired. Cyclic-AMP induces transient blebbing independently of much of the known chemotactic signal transduction machinery, but involving PI3-kinase and downstream PH domain proteins, CRAC and PhdA. Impairment of this PI3-kinase pathway results in slow movement under agarose and cells that produce few blebs, though actin polymerization appears unaffected. We propose that mechanical resistance induces bleb-driven movement in Dictyostelium, which is chemotactic and controlled through PI3-kinase.
Collapse
|
40
|
Fets L, Nichols JME, Kay RR. A PIP5 kinase essential for efficient chemotactic signaling. Curr Biol 2014; 24:415-21. [PMID: 24485835 PMCID: PMC3969243 DOI: 10.1016/j.cub.2013.12.052] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 11/24/2013] [Accepted: 12/20/2013] [Indexed: 12/11/2022]
Abstract
In neutrophils and Dictyostelium, chemoattractant gradients generate directed cell migration by eliciting signaling events that bias intrinsic motility and favor the production and retention of upgradient pseudopods. Phosphoinositides are actively regulated during chemotaxis in these cells, most iconically in the production of PI(3,4,5)P3 gradients within the plasma membrane. Although it is now known that PI(3,4,5)P3 signaling is nonessential for gradient sensing, the role of the related phosphoinositide PI(4,5)P2 is little understood, despite its clear importance in many cell biological processes. We describe here a PIP5 kinase, PikI, which produces PI(4,5)P2 and is essential for efficient chemotaxis of Dictyostelium cells. Without PikI, PI(4,5)P2 levels are reduced by 90%, and while pikI(-) cells move at normal speeds, they are highly disorientated in cAMP gradients. Following chemotactic stimulation, Ras is efficiently activated in pikI(-) cells, yet Ras-dependent responses (including activation of PKB) are severely impaired. PikI is phosphorylated by PKB, and in vitro studies of a phosphomimic mutant suggest that this phosphorylation increases PikI activity. We propose that adequate PI(4,5)P2 levels are required to couple activated Ras to its downstream effectors and that these levels are regulated by PikI, making it a crucial player in gradient sensing.
Collapse
Affiliation(s)
- Louise Fets
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | - John M E Nichols
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Robert R Kay
- Cell Biology Division, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| |
Collapse
|
41
|
Huang CH, Tang M, Shi C, Iglesias PA, Devreotes PN. An excitable signal integrator couples to an idling cytoskeletal oscillator to drive cell migration. Nat Cell Biol 2013; 15:1307-16. [PMID: 24142103 PMCID: PMC3838899 DOI: 10.1038/ncb2859] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 09/11/2013] [Indexed: 12/22/2022]
Abstract
It is generally believed that cytoskeletal activities drive random cell migration while signal transduction events initiated by receptors regulate the cytoskeleton to guide cells. However, we find that the cytoskeletal network, involving Scar/Wave, Arp 2/3, and actin binding proteins, is only capable of generating rapid oscillations and undulations of the cell boundary. The signal transduction network, comprising multiple pathways that include Ras GTPases, PI3K, and Rac GTPases, is required to generate the sustained protrusions of migrating cells. The signal transduction network is excitable, displaying wave propagation, refractoriness, and maximal response to suprathreshold stimuli, even in the absence of the cytoskeleton. We suggest that cell motility results from coupling of “pacemaker” signal transduction and “idling motor” cytoskeletal networks, and various guidance cues that modulate the threshold for triggering signal transduction events are integrated to control the mode and direction of migration.
Collapse
Affiliation(s)
- Chuan-Hsiang Huang
- 1] Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA [2]
| | | | | | | | | |
Collapse
|
42
|
Insall R. The interaction between pseudopods and extracellular signalling during chemotaxis and directed migration. Curr Opin Cell Biol 2013; 25:526-31. [PMID: 23747069 DOI: 10.1016/j.ceb.2013.04.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 04/30/2013] [Indexed: 12/22/2022]
Abstract
Eukaryotic chemotaxis is extremely complex. Cells can sense a wide range of stimuli, and many intracellular pathways are simultaneously involved. Recent genetic analyses of the steps between receptors and cytoskeleton, and how the cell controls actin and pseudopod behaviour, have yielded exciting new data but still no coherent understanding of chemotaxis. However, concentrating on pseudopods themselves and the physical processes that regulate them, rather than the internal signalling pathways, can simplify the data and help resolve the underlying mechanism. Direct action of electric fields and physical forces on cell migration suggest that mechanical forces and force-generating proteins like actin and myosin are centrally important in steering cells during chemotaxis.
Collapse
Affiliation(s)
- Robert Insall
- CRUK Beatson Institute for Cancer Research, Switchback Road, Bearsden, Glasgow G61 1BD, UK.
| |
Collapse
|
43
|
He Y, Li D, Cook SL, Yoon MS, Kapoor A, Rao CV, Kenis PJA, Chen J, Wang F. Mammalian target of rapamycin and Rictor control neutrophil chemotaxis by regulating Rac/Cdc42 activity and the actin cytoskeleton. Mol Biol Cell 2013; 24:3369-80. [PMID: 24006489 PMCID: PMC3814157 DOI: 10.1091/mbc.e13-07-0405] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Rictor, a component of mammalian target of rapamycin complex 2 (mTORC2), controls neutrophil chemotaxis by regulating the dynamics of the actin cytoskeleton via Rac and Cdc42. This function of Rictor is independent of mTORC2 and the kinase activity of mTOR. Chemotaxis allows neutrophils to seek out sites of infection and inflammation. The asymmetric accumulation of filamentous actin (F-actin) at the leading edge provides the driving force for protrusion and is essential for the development and maintenance of neutrophil polarity. The mechanism that governs actin cytoskeleton dynamics and assembly in neutrophils has been extensively explored and is still not fully understood. By using neutrophil-like HL-60 cells, we describe a pivotal role for Rictor, a component of mammalian target of rapamycin complex 2 (mTORC2), in regulating assembly of the actin cytoskeleton during neutrophil chemotaxis. Depletion of mTOR and Rictor, but not Raptor, impairs actin polymerization, leading-edge establishment, and directional migration in neutrophils stimulated with chemoattractants. Of interest, depletion of mSin1, an integral component of mTORC2, causes no detectable defects in neutrophil polarity and chemotaxis. In addition, experiments with chemical inhibition and kinase-dead mutants indicate that mTOR kinase activity and AKT phosphorylation are dispensable for chemotaxis. Instead, our results suggest that the small Rho GTPases Rac and Cdc42 serve as downstream effectors of Rictor to regulate actin assembly and organization in neutrophils. Together our findings reveal an mTORC2- and mTOR kinase–independent function and mechanism of Rictor in the regulation of neutrophil chemotaxis.
Collapse
Affiliation(s)
- Yuan He
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kortholt A, Keizer-Gunnink I, Kataria R, Van Haastert PJM. Ras activation and symmetry breaking during Dictyostelium chemotaxis. J Cell Sci 2013; 126:4502-13. [PMID: 23886948 DOI: 10.1242/jcs.132340] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Central to chemotaxis is the molecular mechanism by which a shallow spatial gradient of chemoattractant induces symmetry breaking of activated signaling molecules. Previously, we have used Dictyostelium mutants to investigate the minimal requirements for chemotaxis, and identified a basal signaling module providing activation of Ras and F-actin at the leading edge. Here, we show that Ras activation after application of a pipette releasing the chemoattractant cAMP has three phases, each depending on specific guanine-nucleotide-exchange factors (GEFs). Initially a transient activation of Ras occurs at the entire cell boundary, which is proportional to the local cAMP concentrations and therefore slightly stronger at the front than in the rear of the cell. This transient Ras activation is present in gα2 (gpbB)-null cells but not in gβ (gpbA)-null cells, suggesting that Gβγ mediates the initial activation of Ras. The second phase is symmetry breaking: Ras is activated only at the side of the cell closest to the pipette. Symmetry breaking absolutely requires Gα2 and Gβγ, but not the cytoskeleton or four cAMP-induced signaling pathways, those dependent on phosphatidylinositol (3,4,5)-triphosphate [PtdIns(3,4,5)P3], cGMP, TorC2 and PLA2. As cells move in the gradient, the crescent of activated Ras in the front half of the cell becomes confined to a small area at the utmost front of the cell. Confinement of Ras activation leads to cell polarization, and depends on cGMP formation, myosin and F-actin. The experiments show that activation, symmetry breaking and confinement of Ras during Dictyostelium chemotaxis uses different G-protein subunits and a multitude of Ras GEFs and GTPase-activating proteins (GAPs).
Collapse
Affiliation(s)
- Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | | | | | | |
Collapse
|
45
|
Jin T. Gradient sensing during chemotaxis. Curr Opin Cell Biol 2013; 25:532-7. [PMID: 23880435 DOI: 10.1016/j.ceb.2013.06.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/18/2013] [Accepted: 06/19/2013] [Indexed: 11/17/2022]
Abstract
Eukaryotic cells have the ability to sense chemoattractant gradients and to migrate toward the sources of attractants. The chemical gradient-guided cell movement is referred to as chemotaxis. Chemoattractants are detected by members of G-protein-coupled receptors (GPCRs) that link to heterotrimeric G-proteins. The GPCR/G-protein sensing machinery is able to translate external chemoattractants fields into intercellular cues, which direct reorganization of the actin cytoskeleton that drives cell movement. Here, I review our current understanding of the formation of chemoattractant gradients in vivo, the GPCR-mediated gradient sensing, and the sophisticated signaling network that guides the function of the actin cytoskeleton.
Collapse
Affiliation(s)
- Tian Jin
- Chemotaxis Signal Section, Laboratory of Immunogenetics, NIAID, NIH, Twinbrook II Facility, 12441 Parklawn Drive, Rockville, MD 20852, United States.
| |
Collapse
|
46
|
Hoeller O, Bolourani P, Clark J, Stephens LR, Hawkins PT, Weiner OD, Weeks G, Kay RR. Two distinct functions for PI3-kinases in macropinocytosis. J Cell Sci 2013; 126:4296-307. [PMID: 23843627 DOI: 10.1242/jcs.134015] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Class-1 PI3-kinases are major regulators of the actin cytoskeleton, whose precise contributions to chemotaxis, phagocytosis and macropinocytosis remain unresolved. We used systematic genetic ablation to examine this question in growing Dictyostelium cells. Mass spectroscopy shows that a quintuple mutant lacking the entire genomic complement of class-1 PI3-kinases retains only 10% of wild-type PtdIns(3,4,5)P3 levels. Chemotaxis to folate and phagocytosis of bacteria proceed normally in the quintuple mutant but macropinocytosis is abolished. In this context PI3-kinases show specialized functions, only one of which is directly linked to gross PtdIns(3,4,5)P3 levels: macropinosomes originate in patches of PtdIns(3,4,5)P3, with associated F-actin-rich ruffles, both of which depend on PI3-kinase 1/2 (PI3K1/2) but not PI3K4, whereas conversion of ruffles into vesicles requires PI3K4. A biosensor derived from the Ras-binding domain of PI3K1 suggests that Ras is activated throughout vesicle formation. Binding assays show that RasG and RasS interact most strongly with PI3K1/2 and PI3K4, and single mutants of either Ras have severe macropinocytosis defects. Thus, the fundamental function of PI3-kinases in growing Dictyostelium cells is in macropinocytosis where they have two distinct functions, supported by at least two separate Ras proteins.
Collapse
Affiliation(s)
- Oliver Hoeller
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Shi C, Huang CH, Devreotes PN, Iglesias PA. Interaction of motility, directional sensing, and polarity modules recreates the behaviors of chemotaxing cells. PLoS Comput Biol 2013; 9:e1003122. [PMID: 23861660 PMCID: PMC3701696 DOI: 10.1371/journal.pcbi.1003122] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 05/16/2013] [Indexed: 02/04/2023] Open
Abstract
Chemotaxis involves the coordinated action of separable but interrelated processes: motility, gradient sensing, and polarization. We have hypothesized that these are mediated by separate modules that account for these processes individually and that, when combined, recreate most of the behaviors of chemotactic cells. Here, we describe a mathematical model where the modules are implemented in terms of reaction-diffusion equations. Migration and the accompanying changes in cellular morphology are demonstrated in simulations using a mechanical model of the cell cortex implemented in the level set framework. The central module is an excitable network that accounts for random migration. The response to combinations of uniform stimuli and gradients is mediated by a local excitation, global inhibition module that biases the direction in which excitability is directed. A polarization module linked to the excitable network through the cytoskeleton allows unstimulated cells to move persistently and, for cells in gradients, to gradually acquire distinct sensitivity between front and back. Finally, by varying the strengths of various feedback loops in the model we obtain cellular behaviors that mirror those of genetically altered cell lines. Chemotaxis is the movement of cells in response to spatial gradients of chemical cues. While single-celled organisms rely on sensing and responding to chemical gradients to search for nutrients, chemotaxis is also an essential component of the mammalian immune system. However, chemotaxis can also be deleterious, since chemotactic tumor cells can lead to metastasis. Due to its importance, understanding the process by which cells sense and respond to chemical gradients has attracted considerable interest. Moreover, because of the complexity of chemotactic signaling, which includes multiple feedback loops and redundant pathways, this has been a research area in which computational models have had a significant impact in understanding experimental findings. Here, we propose a modular description of the signaling network that regulates chemotaxis. The modules describe different processes that are observed in chemotactic cells. In addition to accounting for these behaviors individually, we show that the overall system recreates many features of the directed motion of migrating cells. The signaling described by our modules is implemented as a series of equations, whereas movement and the accompanying cellular deformations are simulated using a mechanical model of the cell and implemented using level set methods, a method that allows simulations of cells as they change morphology.
Collapse
Affiliation(s)
- Changji Shi
- Department of Electrical and Computer Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Chuan-Hsiang Huang
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Peter N. Devreotes
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Pablo A. Iglesias
- Department of Electrical and Computer Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Biological Physics, Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
- * E-mail:
| |
Collapse
|
48
|
Douglass KM, Sparrow NA, Bott M, Fernandez-Valle C, Dogariu A. Measuring anisotropic cell motility on curved substrates. JOURNAL OF BIOPHOTONICS 2013; 6:387-392. [PMID: 22887747 DOI: 10.1002/jbio.201200089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/10/2012] [Accepted: 07/10/2012] [Indexed: 06/01/2023]
Abstract
Schwann cell motility was observed on laminin-coated quartz cylinders with different curvatures over an 18 hour period. A new analysis based on difference images helped to determine the minimal radius of curvature, 46 μm, which restricted motility along the cylinder axis. The migration speed, measured by calculating differences between successive images in the time series, ranged between 0.3 to 0.8 μm per minute and is similar to previously reported rates for Schwann cells. Difference images provide a rapid and simple method for the analysis of cell motility on large populations of cells.
Collapse
Affiliation(s)
- Kyle M Douglass
- CREOL, The College of Optics and Photonics, University of Central Florida, Orlando, FL 32816, USA
| | | | | | | | | |
Collapse
|
49
|
Runne C, Chen S. WD40-repeat proteins control the flow of Gβγ signaling for directional cell migration. Cell Adh Migr 2013; 7:214-8. [PMID: 23302952 DOI: 10.4161/cam.22940] [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: 11/19/2022] Open
Abstract
The ability of cells to generate a highly polarized intracellular signal through G protein-coupled receptors (GPCRs) is essential for their migration toward chemoattractants. The Gβγ subunits of heterotrimeric G proteins play a critical role in transmitting chemotactic signals from GPCRs via the activation of diverse effectors, including PLCβ and PI3K, primarily at the leading edge of cells. Although Gβγ can directly activate many of these effectors through protein-protein interactions in vitro, it remains unclear how Gβγ spatially and temporally orchestrates the activation of these effectors in vivo. A yeast two-hybrid screen for Gβ interacting proteins identified two WD40-repeat domain containing proteins, RACK1 and WDR26, which are predicted to serve as scaffolding/adaptor proteins. Previous data indicates that RACK1 negatively regulates Gβγ-mediated leukocyte migration by inhibiting Gβγ-stimulated PLCβ and PI3K activities. In contrast, recently published work by Sun et al. indicates that WDR26 promotes leukocyte migration by enhancing Gβγ-mediated signal transduction. These findings reveal a novel mechanism regulating Gβγ signaling during chemotaxis, namely through the positive and negative regulation of WDR26 and RACK1 on Gβγ to promote and fine tune Gβγ-mediated effector activation, ultimately governing the ability of cells to polarize and migrate toward a chemoattractant gradient.
Collapse
Affiliation(s)
- Caitlin Runne
- Department of Pharmacology; Roy J. and Lucille A. Carver College of Medicine; University of Iowa; Iowa City, IA USA
| | | |
Collapse
|
50
|
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: 178] [Impact Index Per Article: 13.7] [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
|