Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 337
Filter
1.
Int J Mol Sci ; 23(2)2022 Jan 11.
Article in English | MEDLINE | ID: mdl-35054941

ABSTRACT

Clostridium botulinum produces the botulinum neurotoxin that causes botulism, a rare but potentially lethal paralysis. Endospores play an important role in the survival, transmission, and pathogenesis of C. botulinum. C. botulinum strains are very diverse, both genetically and ecologically. Group I strains are terrestrial, mesophilic, and produce highly heat-resistant spores, while Group II strains can be terrestrial (type B) or aquatic (type E) and are generally psychrotrophic and produce spores of moderate heat resistance. Group III strains are either terrestrial or aquatic, mesophilic or slightly thermophilic, and the heat resistance properties of their spores are poorly characterized. Here, we analyzed the sporulation dynamics in population, spore morphology, and other spore properties of 10 C. botulinum strains belonging to Groups I-III. We propose two distinct sporulation strategies used by C. botulinum Groups I-III strains, report their spore properties, and suggest a putative role for the exosporium in conferring high heat resistance. Strains within each physiological group produced spores with similar characteristics, likely reflecting adaptation to respective environmental habitats. Our work provides new information on the spores and on the population and single-cell level strategies in the sporulation of C. botulinum.


Subject(s)
Botulism/microbiology , Cell Surface Extensions/physiology , Clostridium botulinum/physiology , Microbial Viability , Spores, Bacterial/physiology , Cell Surface Extensions/ultrastructure , Clostridium botulinum/ultrastructure , Spores, Bacterial/ultrastructure
2.
PLoS Comput Biol ; 17(11): e1009576, 2021 11.
Article in English | MEDLINE | ID: mdl-34748539

ABSTRACT

Advances in genetic engineering technologies have allowed the construction of artificial genetic circuits, which have been used to generate spatial patterns of differential gene expression. However, the question of how cells can be programmed, and how complex the rules need to be, to achieve a desired tissue morphology has received less attention. Here, we address these questions by developing a mathematical model to study how cells can collectively grow into clusters with different structural morphologies by secreting diffusible signals that can influence cellular growth rates. We formulate how growth regulators can be used to control the formation of cellular protrusions and how the range of achievable structures scales with the number of distinct signals. We show that a single growth inhibitor is insufficient for the formation of multiple protrusions but may be achieved with multiple growth inhibitors, and that other types of signals can regulate the shape of protrusion tips. These examples illustrate how our approach could potentially be used to guide the design of regulatory circuits for achieving a desired target structure.


Subject(s)
Cell Proliferation/physiology , Cell Shape/physiology , Cellular Reprogramming Techniques/methods , Models, Biological , Animals , Cell Aggregation/physiology , Cell Communication/physiology , Cell Surface Extensions/physiology , Cellular Reprogramming Techniques/statistics & numerical data , Computational Biology , Computer Simulation , Gene Regulatory Networks , Genetic Engineering/methods , Genetic Engineering/statistics & numerical data , Growth Inhibitors/physiology , Humans , Morphogenesis/physiology , Synthetic Biology
3.
Cell Rep ; 37(7): 110008, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34788623

ABSTRACT

Clathrin-mediated endocytosis (CME) is critical for cellular signal transduction, receptor recycling, and membrane homeostasis in mammalian cells. Acute depletion of cholesterol disrupts CME, motivating analysis of CME dynamics in the context of human disorders of cholesterol metabolism. We report that inhibition of post-squalene cholesterol biosynthesis impairs CME. Imaging of membrane bending dynamics and the CME pit ultrastructure reveals prolonged clathrin pit lifetimes and shallow clathrin-coated structures, suggesting progressive impairment of curvature generation correlates with diminishing sterol abundance. Sterol structural requirements for efficient CME include 3' polar head group and B-ring conformation, resembling the sterol structural prerequisites for tight lipid packing and polarity. Furthermore, Smith-Lemli-Opitz fibroblasts with low cholesterol abundance exhibit deficits in CME-mediated transferrin internalization. We conclude that sterols lower the energetic costs of membrane bending during pit formation and vesicular scission during CME and suggest that reduced CME activity may contribute to cellular phenotypes observed within disorders of cholesterol metabolism.


Subject(s)
Clathrin-Coated Vesicles/metabolism , Endocytosis/physiology , Sterols/pharmacology , Cell Surface Extensions/metabolism , Cell Surface Extensions/physiology , Cholesterol/metabolism , Clathrin/metabolism , Fibroblasts/metabolism , HEK293 Cells , Humans , Lipid Metabolism/physiology , Lipids/physiology , Membrane Proteins/metabolism , Receptors, Transferrin/metabolism , Sterols/metabolism
4.
Appl Opt ; 60(25): G10-G18, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34613190

ABSTRACT

Understanding biological responses to directed energy (DE) is critical to ensure the safety of personnel within the Department of Defense. At the Air Force Research Laboratory, we have developed or adapted advanced optical imaging systems that quantify biophysical responses to DE. One notable cellular response to DE exposure is the formation of blebs, or semi-spherical protrusions of the plasma membrane in living cells. In this work, we demonstrate the capacity of quantitative phase imaging (QPI) to both visualize and quantify the formation of membrane blebs following DE exposure. QPI is an interferometric imaging tool that uses optical path length as a label-free contrast mechanism and is sensitive to the non-aqueous mass density, or dry mass, of living cells. Blebs from both CHO-K1 and U937 cells were generated after exposure to a series of 600 ns, 21.2 kV/cm electric pulses. These blebs were visualized in real time, and their dry mass relative to the rest of the cell body was quantified as a function of time. It is our hope that this system will lead to an improved understanding of both DE-induced and apoptotic blebbing.


Subject(s)
Biophysical Phenomena/physiology , Cell Membrane , Cell Surface Extensions , Microscopy, Interference/methods , Optical Imaging/methods , Animals , CHO Cells , Cell Surface Extensions/physiology , Cell Surface Extensions/ultrastructure , Cricetulus , Electric Stimulation/methods , Equipment Design , Humans , Microscopy, Interference/instrumentation , Optical Imaging/instrumentation , Organelle Size , U937 Cells
5.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504018

ABSTRACT

During activation the platelet cytoskeleton is reorganized, inducing adhesion to the extracellular matrix and cell spreading. These processes are critical for wound healing and clot formation. Initially, this task relies on the formation of strong cellular-extracellular matrix interactions, exposed in subendothelial lesions. Despite the medical relevance of these processes, there is a lack of high-resolution structural information on the platelet cytoskeleton controlling cell spreading and adhesion. Here, we present in situ structural analysis of membrane receptors and the underlying cytoskeleton in platelet protrusions by applying cryoelectron tomography to intact platelets. We utilized three-dimensional averaging procedures to study receptors at the plasma membrane. Analysis of substrate interaction-free receptors yielded one main structural class resolved to 26 Å, resembling the αIIbß3 integrin folded conformation. Furthermore, structural analysis of the actin network in pseudopodia indicates a nonuniform polarity of filaments. This organization would allow generation of the contractile forces required for integrin-mediated cell adhesion.


Subject(s)
Actin Cytoskeleton , Actins/chemistry , Blood Platelets/physiology , Cell Membrane/metabolism , Cell Surface Extensions/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Actins/metabolism , Cell Adhesion , Humans , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
6.
PLoS Comput Biol ; 17(8): e1009237, 2021 08.
Article in English | MEDLINE | ID: mdl-34383753

ABSTRACT

Navigation of fast migrating cells such as amoeba Dictyostelium and immune cells are tightly associated with their morphologies that range from steady polarized forms that support high directionality to those more complex and variable when making frequent turns. Model simulations are essential for quantitative understanding of these features and their origins, however systematic comparisons with real data are underdeveloped. Here, by employing deep-learning-based feature extraction combined with phase-field modeling framework, we show that a low dimensional feature space for 2D migrating cell morphologies obtained from the shape stereotype of keratocytes, Dictyostelium and neutrophils can be fully mapped by an interlinked signaling network of cell-polarization and protrusion dynamics. Our analysis links the data-driven shape analysis to the underlying causalities by identifying key parameters critical for migratory morphologies both normal and aberrant under genetic and pharmacological perturbations. The results underscore the importance of deciphering self-organizing states and their interplay when characterizing morphological phenotypes.


Subject(s)
Cell Movement/physiology , Deep Learning , Models, Biological , Animals , Cell Polarity/physiology , Cell Shape/physiology , Cell Surface Extensions/physiology , Cells, Cultured , Cichlids , Computational Biology , Computer Simulation , Dictyostelium/cytology , Dictyostelium/physiology , Fibroblasts/cytology , Fibroblasts/physiology , HL-60 Cells , Humans
7.
Nat Commun ; 12(1): 1548, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33750780

ABSTRACT

Reconstituting artificial proto-cells capable of transducing extracellular signals into cytoskeletal changes can reveal fundamental principles of how non-equilibrium phenomena in cellular signal transduction affect morphogenesis. Here, we generated a Synthetic Morphogenic Membrane System (SynMMS) by encapsulating a dynamic microtubule (MT) aster and a light-inducible signaling system driven by GTP/ATP chemical potential into cell-sized liposomes. Responding to light cues in analogy to morphogens, this biomimetic design embodies basic principles of localized Rho-GTPase signal transduction that generate an intracellular MT-regulator signaling gradient. Light-induced signaling promotes membrane-deforming growth of MT-filaments by dynamically elevating the membrane-proximal tubulin concentration. The resulting membrane deformations enable recursive coupling of the MT-aster with the signaling system, which generates global self-organized morphologies that reorganize towards local external cues in dependence on prior shape. SynMMS thereby signifies a step towards bio-inspired engineering of self-organized cellular morphogenesis.


Subject(s)
Cues , Liposomes , Morphogenesis/physiology , Artificial Cells , Biophysical Phenomena , Cell Surface Extensions/physiology , Centrosome , Cytoskeleton/metabolism , Humans , Liposomes/chemistry , Microtubules/metabolism , Recombinant Proteins , Signal Transduction , Stathmin/metabolism , Synthetic Biology , Tubulin/metabolism , rho GTP-Binding Proteins/metabolism
8.
Neurochem Res ; 46(10): 2525-2537, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33523395

ABSTRACT

Astroglia are neural cells, heterogeneous in form and function, which act as supportive elements of the central nervous system; astrocytes contribute to all aspects of neural functions in health and disease. Through their highly ramified processes, astrocytes form close physical contacts with synapses and blood vessels, and are integrated into functional syncytia by gap junctions. Astrocytes interact among themselves and with other cells types (e.g., neurons, microglia, blood vessel cells) by an elaborate repertoire of chemical messengers and receptors; astrocytes also influence neural plasticity and synaptic transmission through maintaining homeostasis of neurotransmitters, K+ buffering, synaptic isolation and control over synaptogenesis and synaptic elimination. Satellite glial cells (SGCs) are the most abundant glial cells in sensory ganglia, and are believed to play major roles in sensory functions, but so far research into SGCs attracted relatively little attention. In this review we compare SGCs to astrocytes with the purpose of using the vast knowledge on astrocytes to explore new aspects of SGCs. We survey the main properties of these two cells types and highlight similarities and differences between them. We conclude that despite the much greater diversity in morphology and signaling mechanisms of astrocytes, there are some parallels between them and SGCs. Both types serve as boundary cells, separating different compartments in the nervous system, but much more needs to be learned on this aspect of SGCs. Astrocytes and SGCs employ chemical messengers and calcium waves for intercellular signaling, but their significance is still poorly understood for both cell types. Both types undergo major changes under pathological conditions, which have a protective function, but an also contribute to disease, and chronic pain in particular. The knowledge obtained on astrocytes is likely to benefit future research on SGCs.


Subject(s)
Astrocytes/classification , Astrocytes/physiology , Animals , Astrocytes/cytology , Astrocytes/pathology , Calcium Signaling/physiology , Cell Surface Extensions/physiology , Gap Junctions/physiology , Humans , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology
9.
Open Biol ; 10(8): 200039, 2020 08.
Article in English | MEDLINE | ID: mdl-32810422

ABSTRACT

Understanding the mechanisms of cell-to-cell communication is one of the fundamental questions in biology and medicine. In particular, long-range signalling where cells communicate over several cell diameters is vital during development and homeostasis. The major morphogens, their receptors and intracellular signalling cascades have largely been identified; however, there is a gap in our knowledge of how such signalling factors are propagated over a long distance. In addition to the diffusion-based propagation model, new modalities of disseminating signalling molecules have been identified. It has been shown that cells can communicate with direct contact through long, thin cellular protrusions between signal sending and receiving cells at a distance. Recent studies have revealed a type of cellular protrusion termed 'airinemes' in zebrafish pigment cell types. They share similarities with previously reported cellular protrusions; however, they also exhibit distinct morphology and features. Airinemes are indispensable for pigment pattern development by mediating long-distance Delta-Notch signalling between different pigment cell types. Notably, airineme-mediated signalling is dependent on skin-resident macrophages. Key findings of airineme-mediated intercellular signalling in pattern development, their interplay with macrophages and their implications for the understanding of cellular protrusion-mediated intercellular communication will be discussed.


Subject(s)
Cell Communication , Cell Surface Extensions/physiology , Macrophages/physiology , Signal Transduction , Animals , Biological Transport , Cell Shape , Cytoplasmic Vesicles/metabolism , Humans , Organ Specificity
10.
mBio ; 11(4)2020 08 04.
Article in English | MEDLINE | ID: mdl-32753489

ABSTRACT

Invasion of the colon wall by Entamoeba histolytica during amoebic dysentery entails migration of trophozoites through tissue layers that are rich in extracellular matrix. Transcriptional silencing of the E. histolytica surface metalloprotease EhMSP-1 produces hyperadherent less-motile trophozoites that are deficient in forming invadosomes. Reversible protein phosphorylation is often implicated in regulation of cell motility and invadosome formation. To identify such intermediaries of the EhMSP-1-silenced phenotype, here we compared the phosphoproteomes of EhMSP-1-silenced and vector control trophozoites by using quantitative tandem mass spectrometry-based proteomics. Six proteins were found to be differentially phosphorylated in EhMSP-1-silenced and control cells, including EhCoactosin, a member of the ADF/cofilin family of actin-binding proteins, which was more frequently phosphorylated at serine 147. Regulated overexpression of wild-type, phosphomimetic, and nonphosphorylatable EhCoactosin variants was used to test if phosphorylation functions in control of E. histolytica actin dynamics. Each of the overexpressed proteins colocalized with F-actin during E. histolytica phagocytosis. Nonetheless, trophozoites overexpressing an EhCoactosin phosphomimetic mutant formed more and poorly coordinated cell membrane protrusions compared to those in control or cells expressing a nonphosphorylatable mutant, while trophozoites overexpressing nonphosphorylatable EhCoactosin were significantly more motile within a model of mammalian extracellular matrix. Therefore, although EhCoactosin's actin-binding ability appeared unaffected by phosphorylation, EhCoactosin phosphorylation helps to regulate amoebic motility. These data help to understand the mechanisms underlying altered adherence and motility in EhMSP-1-silenced trophozoites and lay the groundwork for identifying kinases and phosphatases critical for control of amoebic invasiveness.IMPORTANCE Invasive amoebiasis, caused by the intestinal parasite Entamoeba histolytica, causes life-threatening diarrhea and liver abscesses, but, for unknown reasons, only approximately 10% of E. histolytica infections become symptomatic. A key requirement of invasion is the ability of the parasite to migrate through tissue layers. Here, we systematically looked for differences in protein phosphorylation between control parasites and a previously identified hyperadherent E. histolytica cell line that has reduced motility. We identified EhCoactosin, an actin-binding protein not previously known to be phosphoregulated, as one of the differentially phosphorylated proteins in E. histolytica and demonstrated that EhCoactosin phosphorylation functions in control of cell membrane dynamics and amoebic motility. This and the additional differentially phosphorylated proteins reported lay the groundwork for identifying kinases and phosphatases that regulate tissue invasiveness.


Subject(s)
Cell Surface Extensions/physiology , Entamoeba histolytica/metabolism , Microfilament Proteins/metabolism , Protozoan Proteins/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Entamoeba histolytica/genetics , Movement , Phagocytosis , Phosphoproteins/genetics , Phosphorylation , Proteomics , Protozoan Proteins/genetics
11.
Front Immunol ; 11: 1097, 2020.
Article in English | MEDLINE | ID: mdl-32595635

ABSTRACT

Phagocytosis is a specialized process that enables cellular ingestion and clearance of microbes, dead cells and tissue debris that are too large for other endocytic routes. As such, it is an essential component of tissue homeostasis and the innate immune response, and also provides a link to the adaptive immune response. However, ingestion of large particulate materials represents a monumental task for phagocytic cells. It requires profound reorganization of the cell morphology around the target in a controlled manner, which is limited by biophysical constraints. Experimental and theoretical studies have identified critical aspects associated with the interconnected biophysical properties of the receptors, the membrane, and the actin cytoskeleton that can determine the success of large particle internalization. In this review, we will discuss the major physical constraints involved in the formation of a phagosome. Focusing on two of the most-studied types of phagocytic receptors, the Fcγ receptors and the complement receptor 3 (αMß2 integrin), we will describe the complex molecular mechanisms employed by phagocytes to overcome these physical constraints.


Subject(s)
Phagocytosis/immunology , Phagocytosis/physiology , Actin Cytoskeleton/metabolism , Animals , Biophysical Phenomena , Cell Movement/immunology , Cell Movement/physiology , Cell Surface Extensions/immunology , Cell Surface Extensions/physiology , Humans , Ligands , Macrophage-1 Antigen/chemistry , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/physiology , Models, Immunological , Myosin Type II/immunology , Myosin Type II/physiology , Phagosomes/immunology , Phagosomes/physiology , Protein Conformation , Pseudopodia/immunology , Pseudopodia/physiology , Receptors, IgG/chemistry , Receptors, IgG/immunology , Receptors, IgG/physiology
12.
Elife ; 92020 06 04.
Article in English | MEDLINE | ID: mdl-32496193

ABSTRACT

Blood vessels (BVs) are considered an integral component of neural stem cells (NSCs) niches. NSCs in the dentate gyrus (DG(have enigmatic elaborated apical cellular processes that are associated with BVs. Whether this contact serves as a mechanism for delivering circulating molecules is not known. Here we uncovered a previously unrecognized communication route allowing exclusive direct access of blood-borne substances to hippocampal NSCs. BBB-impermeable fluorescent tracer injected transcardially to mice is selectively uptaken by DG NSCs within a minute, via the vessel-associated apical processes. These processes, measured >30 nm in diameter, establish direct membrane-to-membrane contact with endothelial cells in specialized areas of irregular endothelial basement membrane and enriched with vesicular activity. Doxorubicin, a brain-impermeable chemotherapeutic agent, is also readily and selectively uptaken by NSCs and reduces their proliferation, which might explain its problematic anti-neurogenic or cognitive side-effect. The newly-discovered NSC-BV communication route explains how circulatory neurogenic mediators are 'sensed' by NSCs.


Subject(s)
Endothelial Cells/cytology , Hippocampus/cytology , Neural Stem Cells/physiology , Animals , Antibiotics, Antineoplastic/metabolism , Basement Membrane/cytology , Basement Membrane/metabolism , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Cell Communication , Cell Surface Extensions/metabolism , Cell Surface Extensions/physiology , Cytoplasmic Vesicles/metabolism , Doxorubicin/metabolism , Endothelial Cells/metabolism , Female , Growth Substances/metabolism , Male , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis
13.
PLoS Genet ; 16(3): e1008694, 2020 03.
Article in English | MEDLINE | ID: mdl-32196488

ABSTRACT

Cell motility is governed by cooperation between the Arp2/3 complex and nucleation-promoting factors from the Wiskott-Aldrich Syndrome Protein (WASP) family, which together assemble actin filament networks to drive membrane protrusion. Here we identify WHIMP (WAVE Homology In Membrane Protrusions) as a new member of the WASP family. The Whimp gene is encoded on the X chromosome of a subset of mammals, including mice. Murine WHIMP promotes Arp2/3-dependent actin assembly, but is less potent than other nucleation factors. Nevertheless, WHIMP-mediated Arp2/3 activation enhances both plasma membrane ruffling and wound healing migration, whereas WHIMP depletion impairs protrusion and slows motility. WHIMP expression also increases Src-family kinase activity, and WHIMP-induced ruffles contain the additional nucleation-promoting factors WAVE1, WAVE2, and N-WASP, but not JMY or WASH. Perturbing the function of Src-family kinases, WAVE proteins, or Arp2/3 complex inhibits WHIMP-driven ruffling. These results suggest that WHIMP-associated actin assembly plays a direct role in membrane protrusion, but also results in feedback control of tyrosine kinase signaling to modulate the activation of multiple WASP-family members.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Movement/physiology , Cell Surface Extensions/physiology , Wiskott-Aldrich Syndrome Protein/metabolism , src-Family Kinases/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Animals , Cell Line , Endocytosis/physiology , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Protein Domains , Signal Transduction , Wiskott-Aldrich Syndrome Protein Family/metabolism
14.
J Bacteriol ; 202(8)2020 03 26.
Article in English | MEDLINE | ID: mdl-32041794

ABSTRACT

The mechanism underlying Spiroplasma swimming is an enigma. This small bacterium possesses two helical shapes with opposite-handedness at a time, and the boundary between them, called a kink, travels down, possibly accompanying the dual rotations of these physically connected helical structures, without any rotary motors such as flagella. Although the outline of dynamics and structural basis has been proposed, the underlying cause to explain the kink translation is missing. We here demonstrated that the cell morphology of Spiroplasma eriocheiris was fixed at the right-handed helix after motility was stopped by the addition of carbonyl cyanide 3-chlorophenylhydrazone (CCCP), and the preferential state was transformed to the other-handedness by the trigger of light irradiation. This process coupled with the generation and propagation of the artificial kink, presumably without any energy input through biological motors. These findings indicate that the coexistence of two chiral helices is sufficient to propagate the kink and thus to propel the cell body.IMPORTANCE Many swimming bacteria generate a propulsion force by rotating helical filaments like a propeller. However, the nonflagellated bacteria Spiroplasma spp. swim without the use of the appendages. The tiny wall-less bacteria possess two chiral helices at a time, and the boundary called a kink travels down, possibly accompanying the dual rotations of the helices. To solve this enigma, we developed an assay to determine the handedness of the body helices at the single-wind level, and demonstrated that the coexistence of body helices triggers the translation of the kink and that the cell body moves by the resultant cell bend propagation. This finding provides us a totally new aspect of bacterial motility, where the body functions as a transformable screw to propel itself forward.


Subject(s)
Cell Surface Extensions/physiology , Spiroplasma/cytology , Biomechanical Phenomena , Cell Polarity , Cell Surface Extensions/chemistry , Models, Biological , Spiroplasma/chemistry , Spiroplasma/physiology
15.
J Neurosci ; 40(7): 1373-1388, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31896671

ABSTRACT

Microglia exhibit multiple, phenotype-dependent motility patterns often triggered by purinergic stimuli. However, little data exist on motility of human microglia in pathological situations. Here we examine motility of microglia stained with a fluorescent lectin in tissue slices from female and male epileptic patients diagnosed with mesial temporal lobe epilepsy or cortical glioma (peritumoral cortex). Microglial shape varied from ramified to amoeboid cells predominantly in regions of high neuronal loss or closer to a tumor. Live imaging revealed unstimulated or purine-induced microglial motilities, including surveillance movements, membrane ruffling, and process extension or retraction. At different concentrations, ADP triggered opposing motilities. Low doses triggered process extension. It was suppressed by P2Y12 receptor antagonists, which also reduced process length and surveillance movements. Higher purine doses caused process retraction and membrane ruffling, which were blocked by joint application of P2Y1 and P2Y13 receptor antagonists. Purinergic effects on motility were similar for all microglia tested. Both amoeboid and ramified cells from mesial temporal lobe epilepsy or peritumoral cortex tissue expressed P2Y12 receptors. A minority of microglia expressed the adenosine A2A receptor, which has been linked with process withdrawal of rodent cells. Laser-mediated tissue damage let us test the functional significance of these effects. Moderate damage induced microglial process extension, which was blocked by P2Y12 receptor antagonists. Overall, the purine-induced motility of human microglia in epileptic tissue is similar to that of rodent microglia in that the P2Y12 receptor initiates process extension. It differs in that retraction is triggered by joint activation of P2Y1/P2Y13 receptors.SIGNIFICANCE STATEMENT Microglial cells are brain-resident immune cells with multiple functions in healthy or diseased brains. These diverse functions are associated with distinct phenotypes, including different microglial shapes. In the rodent, purinergic signaling is associated with changes in cell shape, such as process extension toward tissue damage. However, there are little data on living human microglia, especially in diseased states. We developed a reliable technique to stain microglia from epileptic and glioma patients to examine responses to purines. Low-intensity purinergic stimuli induced process extension, as in rodents. In contrast, high-intensity stimuli triggered a process withdrawal mediated by both P2Y1 and P2Y13 receptors. P2Y1/P2Y13 receptor activation has not previously been linked to microglial morphological changes.


Subject(s)
Epilepsy, Temporal Lobe/physiopathology , Glioma/physiopathology , Microglia/physiology , Receptors, Purinergic P2Y12/physiology , Receptors, Purinergic P2Y1/physiology , Receptors, Purinergic P2/physiology , Supratentorial Neoplasms/physiopathology , Adenosine Diphosphate/pharmacology , Adult , Cell Movement/drug effects , Cell Movement/physiology , Cell Shape/drug effects , Cell Surface Extensions/drug effects , Cell Surface Extensions/physiology , Cell Surface Extensions/ultrastructure , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/pathology , Female , Glioma/pathology , Humans , Intravital Microscopy , Male , Microglia/drug effects , Microglia/ultrastructure , Middle Aged , Plant Lectins , Purinergic Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Supratentorial Neoplasms/pathology , Tuberous Sclerosis/complications
17.
Dev Cell ; 51(4): 460-475.e10, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31607653

ABSTRACT

In development, wound healing, and cancer metastasis, vertebrate cells move through 3D interstitial matrix, responding to chemical and physical guidance cues. Protrusion at the cell front has been extensively studied, but the retraction phase of the migration cycle is not well understood. Here, we show that fast-moving cells guided by matrix cues establish positive feedback control of rear retraction by sensing membrane tension. We reveal a mechanism of rear retraction in 3D matrix and durotaxis controlled by caveolae, which form in response to low membrane tension at the cell rear. Caveolae activate RhoA-ROCK1/PKN2 signaling via the RhoA guanidine nucleotide exchange factor (GEF) Ect2 to control local F-actin organization and contractility in this subcellular region and promote translocation of the cell rear. A positive feedback loop between cytoskeletal signaling and membrane tension leads to rapid retraction to complete the migration cycle in fast-moving cells, providing directional memory to drive persistent cell migration in complex matrices.


Subject(s)
Cell Movement/physiology , Pseudopodia/physiology , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Caveolae/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/physiology , Cell Polarity/physiology , Cell Surface Extensions/metabolism , Cell Surface Extensions/physiology , Cytoskeleton/metabolism , Cytosol/metabolism , Extracellular Matrix/metabolism , Humans , Mice , Protein Kinase C/metabolism , Pseudopodia/metabolism , Rats , Signal Transduction , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
18.
Essays Biochem ; 63(5): 595-606, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31324705

ABSTRACT

Cell migration is a fundamental biological process involved in tissue formation and homeostasis. The correct polarization of motile cells is critical to ensure directed movement, and is orchestrated by many intrinsic and extrinsic factors. Of these, the subcellular distribution of mRNAs and the consequent spatial control of translation are key modulators of cell polarity. mRNA transport is dependent on cis-regulatory elements within transcripts, which are recognized by trans-acting proteins that ensure the efficient delivery of certain messages to the leading edge of migrating cells. At their destination, translation of localized mRNAs then participates in regional cellular responses underlying cell motility. In this review, we summarize the key findings that established mRNA targetting as a critical driver of cell migration and how the characterization of polarized mRNAs in motile cells has been expanded from just a few species to hundreds of transcripts. We also describe the molecular control of mRNA trafficking, subsequent mechanisms of local protein synthesis and how these ultimately regulate cell polarity during migration.


Subject(s)
Cell Movement/physiology , RNA, Messenger/metabolism , Actins/metabolism , Animals , Cell Surface Extensions/physiology , Humans , Microtubules/metabolism , Protein Biosynthesis/physiology , RNA Transport/physiology
19.
PLoS Biol ; 17(6): e3000060, 2019 06.
Article in English | MEDLINE | ID: mdl-31233488

ABSTRACT

Apicomplexan parasites invade host cells in an active process involving their ability to move by gliding motility. While the acto-myosin system of the parasite plays a crucial role in the formation and release of attachment sites during this process, there are still open questions regarding the involvement of other mechanisms in parasite motility. In many eukaryotes, a secretory-endocytic cycle leads to the recycling of receptors (integrins), necessary to form attachment sites, regulation of surface area during motility, and generation of retrograde membrane flow. Here, we demonstrate that endocytosis operates during gliding motility in Toxoplasma gondii and appears to be crucial for the establishment of retrograde membrane flow, because inhibition of endocytosis blocks retrograde flow and motility. We demonstrate that extracellular parasites can efficiently incorporate exogenous material, such as labelled phospholipids, nanogold particles (NGPs), antibodies, and Concanavalin A (ConA). Using labelled phospholipids, we observed that the endocytic and secretory pathways of the parasite converge, and endocytosed lipids are subsequently secreted, demonstrating the operation of an endocytic-secretory cycle. Together our data consolidate previous findings, and we propose an additional model, working in parallel to the acto-myosin motor, that reconciles parasite motility with observations in other eukaryotes: an apicomplexan fountain-flow-model for parasite motility.


Subject(s)
Cell Movement/physiology , Endocytosis/physiology , Toxoplasma/metabolism , Actins/metabolism , Animals , Cell Adhesion/physiology , Cell Surface Extensions/physiology , Membrane Proteins/metabolism , Myosins/metabolism , Parasites , Protozoan Proteins/metabolism , Secretory Pathway/physiology , Toxoplasma/physiology
20.
Dev Cell ; 49(2): 189-205.e6, 2019 04 22.
Article in English | MEDLINE | ID: mdl-31014479

ABSTRACT

Efficient chemotaxis requires rapid coordination between different parts of the cell in response to changing directional cues. Here, we investigate the mechanism of front-rear coordination in chemotactic neutrophils. We find that changes in the protrusion rate at the cell front are instantaneously coupled to changes in retraction at the cell rear, while myosin II accumulation at the rear exhibits a reproducible 9-15-s lag. In turning cells, myosin II exhibits dynamic side-to-side relocalization at the cell rear in response to turning of the leading edge and facilitates efficient turning by rapidly re-orienting the rear. These manifestations of front-rear coupling can be explained by a simple quantitative model incorporating reversible actin-myosin interactions with a rearward-flowing actin network. Finally, the system can be tuned by the degree of myosin regulatory light chain (MRLC) phosphorylation, which appears to be set in an optimal range to balance persistence of movement and turning ability.


Subject(s)
Chemotaxis/physiology , Myosin Type II/physiology , Neutrophils/physiology , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Animals, Genetically Modified , Cell Line , Cell Movement/physiology , Cell Polarity/physiology , Cell Surface Extensions/physiology , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Female , Humans , Myosin Type II/metabolism , Myosins/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL