Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.705
Filter
1.
Sci Signal ; 16(805): eadd1845, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37788324

ABSTRACT

Human neutrophils respond to multiple chemoattractants to guide their migration from the vasculature to sites of infection and injury, where they clear pathogens and amplify inflammation. To properly focus their responses during this complex navigation, neutrophils prioritize pathogen- and injury-derived signals over long-range inflammatory signals, such as the leukotriene LTB4, secreted by host cells. Different chemoattractants can also drive qualitatively different modes of migration even though their receptors couple to the same Gαi family of G proteins. Here, we used live-cell imaging to demonstrate that the responses differed in their signaling dynamics. Low-priority chemoattractants caused transient responses, whereas responses to high-priority chemoattractants were sustained. We observed this difference in both primary neutrophils and differentiated HL-60 cells, for downstream signaling mediated by Ca2+, a major regulator of secretion, and Cdc42, a primary regulator of polarity and cell steering. The rapid attenuation of Cdc42 activation in response to LTB4 depended on the phosphorylation sites Thr308 and Ser310 in the carboxyl-terminal tail of its receptor LTB4R in a manner independent of endocytosis. Mutation of these residues to alanine impaired chemoattractant prioritization, although it did not affect chemoattractant-dependent differences in migration persistence. Our results indicate that distinct temporal regulation of shared signaling pathways distinguishes between receptors and contributes to chemoattractant prioritization.


Subject(s)
Leukotriene B4 , Neutrophils , Humans , Neutrophils/metabolism , Leukotriene B4/pharmacology , Leukotriene B4/metabolism , Chemotactic Factors/pharmacology , Chemotactic Factors/metabolism , Interleukin-8/metabolism , Signal Transduction
2.
Curr Biol ; 33(9): 1704-1715.e3, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37001521

ABSTRACT

Negative chemotaxis, where eukaryotic cells migrate away from repellents, is important throughout biology, for example, in nervous system patterning and resolution of inflammation. However, the mechanisms by which molecules repel migrating cells are unknown. Here, we use predictive modeling and experiments with Dictyostelium cells to show that competition between different ligands that bind to the same receptor leads to effective chemorepulsion. 8-CPT-cAMP, widely described as a simple chemorepellent, is inactive on its own and only repels cells when it acts in combination with the attractant cAMP. If cells degrade either competing ligand, the pattern of migration becomes more complex; cells may be repelled in one part of a gradient but attracted elsewhere, leading to populations moving in different directions in the same assay or converging in an arbitrary place. More counterintuitively still, two chemicals that normally attract cells can become repellent when combined. Computational models of chemotaxis are now accurate enough to predict phenomena that have not been anticipated by experiments. We have used them to identify new mechanisms that drive reverse chemotaxis, which we have confirmed through experiments with real cells. These findings are important whenever multiple ligands compete for the same receptors.


Subject(s)
Chemotaxis , Dictyostelium , Chemotaxis/physiology , Chemotactic Factors/pharmacology , Chemotactic Factors/metabolism , Dictyostelium/metabolism , Eukaryotic Cells/metabolism
3.
Arterioscler Thromb Vasc Biol ; 43(3): 456-473, 2023 03.
Article in English | MEDLINE | ID: mdl-36700427

ABSTRACT

BACKGROUND: Late vein graft failure is caused by intimal thickening resulting from endothelial cell (EC) damage and inflammation which promotes vascular smooth muscle cell (VSMC) dedifferentiation, migration, and proliferation. Nonphosphorylatable PRH (proline-rich homeodomain) S163C:S177C offers enhanced stability and sustained antimitotic effect. Therefore, we investigated whether adenovirus-delivered PRH S163C:S177C protein attenuates intimal thickening via VSMC phenotype modification without detrimental effects on ECs. METHODS: PRH S163C:S177C was expressed in vitro (human saphenous vein-VSMCs and human saphenous vein-ECs) and in vivo (ligated mouse carotid arteries) by adenoviruses. Proliferation, migration, and apoptosis were quantified and phenotype was assessed using Western blotting for contractile filament proteins and collagen gel contraction. EC inflammation was quantified using VCAM (vascular cell adhesion protein)-1, ICAM (intercellular adhesion molecule)-1, interleukin-6, and monocyte chemotactic factor-1 measurement and monocyte adhesion. Next Generation Sequencing was utilized to identify novel downstream mediators of PRH action and these and intimal thickening were investigated in vivo. RESULTS: PRH S163C:S177C inhibited proliferation, migration, and apoptosis and promoted contractile phenotype (enhanced contractile filament proteins and collagen gel contraction) compared with virus control in human saphenous vein-VSMCs. PRH S163C:S177C expression in human saphenous vein-ECs significantly reduced apoptosis, without affecting cell proliferation and migration, while reducing TNF (tumor necrosis factor)-α-induced VCAM-1 and ICAM-1 and monocyte adhesion and suppressing interleukin-6 and monocyte chemotactic factor-1 protein levels. PRH S163C:S177C expression in ligated murine carotid arteries significantly impaired carotid artery ligation-induced neointimal proliferation and thickening without reducing endothelial coverage. Next Generation Sequencing revealed STAT-1 (signal transducer and activator of transcription 1) and HDAC-9 (histone deacetylase 9) as mediators of PRH action and was supported by in vitro and in vivo analyses. CONCLUSIONS: We observed PRH S163C:S177C attenuated VSMC proliferation, and migration and enhanced VSMC differentiation at least in part via STAT-1 and HDAC-9 signaling while promoting endothelial repair and anti-inflammatory properties. These findings highlight the potential for PRH S163C:S177C to preserve endothelial function whilst suppressing intimal thickening, and reducing late vein graft failure.


Subject(s)
Interleukin-6 , Tunica Intima , Mice , Animals , Humans , Interleukin-6/metabolism , Tunica Intima/pathology , Cell Proliferation , Neointima/pathology , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , Myocytes, Smooth Muscle/metabolism , Cell Movement
4.
Arch Biochem Biophys ; 734: 109497, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36574914

ABSTRACT

NFIC is a potent transcriptional factor involved in many physiological and pathological processes, including tumorigenesis. However, the role of NFIC1, the longest isoform of NFIC, in the progression of triple negative breast cancer (TNBC) remains elusive. Our study demonstrates that overexpression of NFIC1 inhibits the migration and invasion of TNBC MDA-MB-231 cells. NFIC1 regulates the expression of S100A2, and knockdown of S100A2 reverses the inhibitive effects of NFIC1 on the migration and invasion of MDA-MB-231 cells. Furthermore, knockdown of S100A2 activates the MEK/ERK signaling transduction pathway that is inhibited by NFIC1 overexperssion. Treatment with MEK/ERK pathway inhibitor, U0126, abolishes the effects of S100A2 knockdown. In addition, overexpression of NFIC1 in MDA-MB-231 cells increases the expression of epithelial markers and decreases the expression of mesenchymal markers, and these effects could also be reversed by knockdown of S100A2. Collectively, these results demonstrate that NFIC1 inhibits the Epithelial-mesenchymal transition (EMT) of MDA-MB-231 cells by regulating S100A2 expression, which suppress the activation of MEK/ERK pathway. Therefore, our study confirms the role of NFIC1 as a tumor repressor in TNBC, and reveals the molecular mechanism through which NFIC1 inhibits the migration and invasion of MDA-MB-231 cells.


Subject(s)
MAP Kinase Signaling System , Triple Negative Breast Neoplasms , Humans , MDA-MB-231 Cells , Cell Proliferation , Cell Movement , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/pharmacology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , S100 Proteins/metabolism , S100 Proteins/pharmacology
5.
Front Immunol ; 13: 1020117, 2022.
Article in English | MEDLINE | ID: mdl-36341344

ABSTRACT

Chemotaxis plays an essential role in recruitment of leukocytes to sites of inflammation. Eukaryotic cells sense chemoattractant with G protein-coupled receptors (GPCRs) and chemotax toward gradients with an enormous concentration range through adaptation. Cells in adaptation no longer respond to the present stimulus but remain sensitive to stronger stimuli. Thus, adaptation provides a fundamental strategy for eukaryotic cells to chemotax through a gradient. Ras activation is the first step in the chemosensing GPCR signaling pathways that displays a transient activation behavior in both model organism Dictyostelium discoideum and mammalian neutrophils. Recently, it has been revealed that C2GAP1 and CAPRI control the GPCR-mediated adaptation in D. discoideum and human neutrophils, respectively. More importantly, both Ras inhibitors regulate the sensitivity of the cells. These findings suggest an evolutionarily conserved molecular mechanism by which eukaryotic cells gate concentration range of chemoattractants for chemotaxis.


Subject(s)
Chemotaxis , Dictyostelium , Animals , Humans , Chemotaxis/physiology , Dictyostelium/metabolism , Chemotactic Factors/pharmacology , Chemotactic Factors/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Mammals/metabolism
6.
Eur J Pharmacol ; 933: 175267, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36122756

ABSTRACT

The ongoing COVID-19 pandemic is still a challenging problem in the case of infection treatment. The immunomodulatory effect of Nanocurcumin was investigated in the present study in an attempt to counterbalance the immune response and improve the patients' clinical symptoms. 60 confirmed COVID-19 patients and 60 healthy controls enrolled in the study. COVID-19 patients were divided into Nanocurcumin and placebo received groups. Due to the importance of the role of NK cells in this disease, the frequency, cytotoxicity, receptor gene expression of NK cells, and serum secretion levels of inflammatory cytokines IL-1ß, IL-6, TNF-α, as well as circulating C5a as a chemotactic factor an inflammatory mediator was evaluated by flow cytometry, real-time PCR and enzyme-linked immunosorbent assay in both experimental groups before and after the intervention. Given the role of measured factors in the progression and pathogenesis of COVID-19 disease, the results can help find appropriate treatments. The results of this study indicated that the Nanocurcumin could significantly increase the frequency and function of NK cells compared to the placebo-treated group. As an immunomodulatory agent, Nanocurcumin may be a helpful choice to improve NK cell function in COVID-19 patients and improve the clinical outcome of patients.


Subject(s)
COVID-19 Drug Treatment , Case-Control Studies , Chemotactic Factors/pharmacology , Cytokines/metabolism , Humans , Immunity , Inflammation Mediators/pharmacology , Interleukin-6 , Killer Cells, Natural , Pandemics , Tumor Necrosis Factor-alpha/metabolism
7.
Int J Mol Sci ; 23(18)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36142256

ABSTRACT

BACKGROUND: We conducted intratracheal instillations of different molecular weights of polyacrylic acid (PAA) into rats in order to examine what kinds of physicochemical characteristics of acrylic acid-based polymer affect responses in the lung. METHODS: F344 rats were intratracheally exposed to a high molecular weight (HMW) of 598 thousand g/mol or a low molecular weight (LMW) of 30.9 thousand g/mol PAA at low and high doses. Rats were sacrificed at 3 days, 1 week, 1 month, 3 months and 6 months post exposure. RESULTS: HMW PAA caused persistent increases in neutrophil influx, cytokine-induced neutrophil chemoattractants (CINC) in the bronchoalveolar lavage fluid (BALF), and heme oxygenase-1 (HO-1) in the lung tissue from 3 days to 3 months and 6 months following instillation. On the other hand, LMW PAA caused only transient increases in neutrophil influx, CINC in BALF, and HO-1 in the lung tissue from 3 days to up to 1 week or 1 month following instillation. Histopathological findings of the lungs demonstrated that the extensive inflammation and fibrotic changes caused by the HMW PAA was greater than that in exposure to the LMW PAA during the observation period. CONCLUSION: HMW PAA induced persistence of lung disorder, suggesting that molecular weight is a physicochemical characteristic of PAA-induced lung disorder.


Subject(s)
Heme Oxygenase-1 , Lung , Acrylic Resins/pharmacology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Chemotactic Factors/pharmacology , Cytokines/pharmacology , Intubation, Intratracheal , Lung/pathology , Molecular Weight , Rats , Rats, Inbred F344
8.
J Cell Sci ; 135(16)2022 08 15.
Article in English | MEDLINE | ID: mdl-35916164

ABSTRACT

The Dictyostelium atypical mitogen-activated protein kinase (MAPK) Erk2 is required for chemotactic responses to cAMP as amoeba undergo multicellular development. In this study, Erk2 was found to be essential for the cAMP-stimulated translocation of the GATA transcription factor GtaC as indicated by the distribution of a GFP-GtaC reporter. Erk2 was also found to be essential for the translocation of GtaC in response to external folate, a foraging signal that directs the chemotaxis of amoeba to bacteria. Erk1, the only other Dictyostelium MAPK, was not required for the GtaC translocation to either chemoattractant, indicating that GFP-GtaC is a kinase translocation reporter specific for atypical MAPKs. The translocation of GFP-GtaC in response to folate was absent in mutants lacking the folate receptor Far1 or the coupled G-protein subunit Gα4. Loss of GtaC function resulted in enhanced chemotactic movement to folate, suggesting that GtaC suppresses responses to folate. The alteration of four Erk2-preferred phosphorylation sites in GtaC impacted the translocation of GFP-GtaC in response to folate and the GFP-GtaC-mediated rescue of aggregation and development of gtaC- cells. The ability of different chemoattractants to stimulate Erk2-regulated GtaC translocation suggests that atypical MAPK-mediated regulation of transcription factors can contribute to different cell fates.


Subject(s)
Dictyostelium , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , Dictyostelium/metabolism , Folic Acid/pharmacology , GATA Transcription Factors/metabolism , Mitogen-Activated Protein Kinases/metabolism
9.
Biophys J ; 121(13): 2557-2567, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35644945

ABSTRACT

Cell migration on an adhesive substrate surface comprises actin-based protrusion at the front and retraction of the tail in combination with coordinated adhesion to, and detachment from, the substrate. To study the effect of cell-to-substrate adhesion on the chemotactic response of Dictyostelium discoideum cells, we exposed the cells to patterned substrate surfaces consisting of adhesive and inert areas, and forced them by a gradient of chemoattractant to enter the border between the two areas. Wild-type as well as myosin II-deficient cells stop at the border of an adhesive area. They do not detach with their rear part, while on the nonadhesive area they protrude pseudopods at their front toward the source of chemoattractant. Avoidance of the nonadhesive area may cause a cell to move in tangential direction relative to the attractant gradient, keeping its tail at the border of the adhesive surface.


Subject(s)
Dictyostelium , Actins/metabolism , Cell Movement/physiology , Chemotactic Factors/pharmacology , Chemotaxis , Myosin Type II/metabolism , Pseudopodia/metabolism
10.
Exp Cell Res ; 418(1): 113218, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35618013

ABSTRACT

Interplay between models and experimental data advances discovery and understanding in biology, particularly when models generate predictions that allow well-designed experiments to distinguish between alternative mechanisms. To illustrate how this feedback between models and experiments can lead to key insights into biological mechanisms, we explore three examples from cellular slime mold chemotaxis. These examples include studies that identified chemotaxis as the primary mechanism behind slime mold aggregation, discovered that cells likely measure chemoattractant gradients by sensing concentration differences across cell length, and tested the role of cell-associated chemoattractant degradation in shaping chemotactic fields. Although each study used a different model class appropriate to their hypotheses - qualitative, mathematical, or simulation-based - these examples all highlight the utility of modeling to formalize assumptions and generate testable predictions. A central element of this framework is the iterative use of models and experiments, specifically: matching experimental designs to the models, revising models based on mismatches with experimental data, and validating critical model assumptions and predictions with experiments. We advocate for continued use of this interplay between models and experiments to advance biological discovery.


Subject(s)
Dictyosteliida , Dictyostelium , Chemotactic Factors/pharmacology , Chemotaxis , Computer Simulation , Models, Biological
11.
J Vis Exp ; (181)2022 03 25.
Article in English | MEDLINE | ID: mdl-35404349

ABSTRACT

Chemotaxis identification is very important for the research and application of rhizosphere growth-promoting bacteria. We established a straightforward method to quickly identify the chemoattractants that could induce the chemotactic movement of rhizosphere growth-promoting bacteria on sterile glass slides via simple steps. Bacteria solution (OD600 = 0.5) and sterile chemoattractant aqueous solution were added dropwise on the glass slide at an interval of 1 cm. An inoculating loop was used to connect the chemoattractant aqueous solution to the bacterial solution. The slide was kept at room temperature for 20 min on the clean bench. Finally, the chemoattractant aqueous solution was collected for bacterial counting and microscopic observation. In this study, through multiple comparisons of experimental results, the method overcame multiple shortcomings of traditional bacterial chemotaxis methods. The method reduced the error of plate counting and shortened the experimental cycle. For the identification of chemoattractant substances, this new method can save 2-3 days compared to the traditional method. Additionally, this method allows any researcher to systematically complete a bacterial chemotaxis experiment within 1-2 days. The protocol can be considered a valuable resource for understanding plant-microbe interactions.


Subject(s)
Chemotactic Factors , Chemotaxis , Bacteria , Chemotactic Factors/pharmacology , Exudates and Transudates , Plant Roots/microbiology
12.
Trends Cell Biol ; 32(7): 585-596, 2022 07.
Article in English | MEDLINE | ID: mdl-35351380

ABSTRACT

Chemotaxis, where cell movement is steered by chemical gradients, is a widespread and essential way of organising cell behaviour. But where do the instructions come from - who makes gradients, and how are they controlled? We discuss the emerging concept that chemotactic cells often create attractant gradients at the same time as responding to them. This self-guidance is more robust, works across greater distances, and is more informative about the local environment than passive responses. Several mechanisms can establish autonomous gradients. Best known are self-generated gradients, in which the cells degrade a widespread attractant, but cells also produce repellents and 'relay' by secreting fresh attractant after stimulation. Understanding how cells make and interpret their own chemoattractant gradients is fundamental to understanding the spatial patterns seen in all organisms.


Subject(s)
Chemotactic Factors , Chemotaxis , Cell Movement , Chemotactic Factors/chemistry , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , Chemotaxis/physiology , Humans
13.
Adv Biol (Weinh) ; 6(6): e2101312, 2022 06.
Article in English | MEDLINE | ID: mdl-35347887

ABSTRACT

The crawling behavior of leukocytes is driven by the cell morphology transition, which is a direct manifestation of molecular motor machinery. The topographical anisotropy and mechanical stiffness of the substrates are the main physical cues that affect leukocytes' shape generation and migratory responses. However, their combined effects on the cell morphology and motility have been poorly understood, particularly for neutrophils, which are the fastest reacting leukocytes against infections and wounds. Here, spatiotemporally correlated physical parameters are shown, which determine the neutrophil shape change during migratory processes, in response to surface topography and elasticity. Guided crawling and shape generation of individual neutrophils, activated by a uniform concentration of a chemoattractant, are analyzed by adopting elasticity-tunable micropatterning and live cell imaging techniques. Whole cell-level image analysis is performed based on a planar geometric quantification of cell shape and motility. The findings show that the pattern anisotropy and elastic modulus of the substrate induce synergic effects on the shape anisotropy, deformability, and polarization/alignment of crawling neutrophils. How the morphology-motility relationship is affected by different surface microstructures and stiffness is demonstrated. These results imply that the neutrophil shape-motility correlations can be utilized for controlling the immune cell functions with predefined physical microenvironments.


Subject(s)
Chemotactic Factors , Neutrophils , Cell Movement , Cell Shape , Chemotactic Factors/pharmacology , Elasticity
14.
Eur J Pharm Sci ; 172: 106144, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35158054

ABSTRACT

5-Oxo-6,8,11,14-eicosatetraenoic acid (5-oxo-ETE) is the only product of the proinflammatory 5-lipoxygenase pathway with potent chemoattractant effects for human eosinophils, suggesting an important role in eosinophilic diseases such as asthma. 5-Oxo-ETE, acting through its selective OXE receptor, induces dermal eosinophilia in both humans and monkeys. To block its effects, we designed selective indole-based OXE antagonists containing hexyl (S-230) or phenylhexyl (S-C025 and S-Y048) side chains, which inhibit allergen-induced dermal and pulmonary inflammation in monkeys, suggesting that they may be useful therapeutic agents in humans. In this study we identified two metabolic pathways for the phenylhexyl-containing antagonists in liver microsomes: benzylic and N-methyl hydroxylation, resulting in ω-hydroxy, ω-oxo, and NH-containing products with reduced potencies that were identified by mass spectrometry and comparison with synthetic standards. Products of both pathways were also identified in monkey plasma following oral administration of S-C025 and S-Y025, but were less abundant than the α-hydroxy metabolites that we previously identified. Interestingly, the α-hydroxy compounds were not detected in microsomal incubations, suggesting a different origin. The relative rates of metabolism of these antagonists were S-230 >> S-C025 > S-Y048, which may help to explain the differences in their plasma half-lives (S-230 < S-C025 < S-Y048). In conclusion, S-C025 and S-Y048 are metabolized by liver microsomes by benzylic and N-methyl hydroxylation but not by α-hydroxylation, whereas all three pathways exist in vivo. Addition of a phenyl group to the hexyl side chain of these antagonists dramatically reduced their rates of metabolism, which would explain their prolonged in vivo half-lives.


Subject(s)
Eosinophils , Receptors, Eicosanoid , Animals , Anti-Inflammatory Agents/pharmacology , Chemotactic Factors/pharmacology , Haplorhini/metabolism
15.
J Leukoc Biol ; 111(6): 1175-1184, 2022 06.
Article in English | MEDLINE | ID: mdl-35100458

ABSTRACT

Following injury and infection, neutrophils are guided to the affected site by chemoattractants released from injured tissues and invading microbes. During this process (chemotaxis), neutrophils must integrate multiple chemical signals, while also responding to physical constraints and prioritizing their directional decisions to generate an efficient immune response. In some clinical conditions, human neutrophils appear to lose the ability to chemotax efficiently, which may contribute both directly and indirectly to disease pathology. Here, a range of microfluidic designs is utilized to test the sensitivity of chemotaxing neutrophils to various perturbations, including binary decision-making in the context of channels with different chemoattractant gradients, hydraulic resistance, and angle of approach. Neutrophil migration in long narrow channels and planar environments is measured. Conditions in which neutrophils are significantly more likely to choose paths with the steepest chemoattractant gradient and the most direct approach angle, and find that migration efficiency across planar chambers is inversely correlated with chamber diameter. By sequential measurement of neutrophil binary decision-making to different chemoattractant gradients, or chemotactic index in sequential planar environments, data supporting a model of biased random walk for neutrophil chemotaxis are presented.


Subject(s)
Chemotaxis , Neutrophils , Cell Movement/physiology , Chemotactic Factors/pharmacology , Chemotaxis/physiology , Chemotaxis, Leukocyte/physiology , Humans , Neutrophils/physiology
16.
Mol Biol Cell ; 33(3): ar23, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34910560

ABSTRACT

A GPCR-mediated signaling network enables a chemotactic cell to generate adaptative Ras signaling in response to a large range of concentrations of a chemoattractant. To explore potential regulatory mechanisms of GPCR-controlled Ras signaling in chemosensing, we applied a software package, Simmune, to construct detailed spatiotemporal models simulating responses of the cAR1-mediated Ras signaling network. We first determined the dynamics of G-protein activation and Ras signaling in Dictyostelium cells in response to cAMP stimulations using live-cell imaging and then constructed computation models by incorporating potential mechanisms. Using simulations, we validated the dynamics of signaling events and predicted the dynamic profiles of those events in the cAR1-mediated Ras signaling networks with defective Ras inhibitory mechanisms, such as without RasGAP, with RasGAP overexpression, or with RasGAP hyperactivation. We describe a method of using Simmune to construct spatiotemporal models of a signaling network and run computational simulations without writing mathematical equations. This approach will help biologists to develop and analyze computational models that parallel live-cell experiments.


Subject(s)
Chemotactic Factors , Dictyostelium , Chemotactic Factors/pharmacology , GTPase-Activating Proteins , Signal Transduction/physiology , Systems Analysis
17.
Cells ; 10(12)2021 12 10.
Article in English | MEDLINE | ID: mdl-34943993

ABSTRACT

The lamellipodia and pseudopodia of migrating cells are produced and maintained by the Scar/WAVE complex. Thus, actin-based cell migration is largely controlled through regulation of Scar/WAVE. Here, we report that the Abi subunit-but not Scar-is phosphorylated in response to extracellular signalling in Dictyostelium cells. Like Scar, Abi is phosphorylated after the complex has been activated, implying that Abi phosphorylation modulates pseudopodia, rather than causing new ones to be made. Consistent with this, Scar complex mutants that cannot bind Rac are also not phosphorylated. Several environmental cues also affect Abi phosphorylation-cell-substrate adhesion promotes it and increased extracellular osmolarity diminishes it. Both unphosphorylatable and phosphomimetic Abi efficiently rescue the chemotaxis of Abi KO cells and pseudopodia formation, confirming that Abi phosphorylation is not required for activation or inactivation of the Scar/WAVE complex. However, pseudopodia and Scar patches in the cells with unphosphorylatable Abi protrude for longer, altering pseudopod dynamics and cell speed. Dictyostelium, in which Scar and Abi are both unphosphorylatable, can still form pseudopods, but migrate substantially faster. We conclude that extracellular signals and environmental responses modulate cell migration by tuning the behaviour of the Scar/WAVE complex after it has been activated.


Subject(s)
Dictyostelium/metabolism , Extracellular Space/metabolism , Protozoan Proteins/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Chemotactic Factors/pharmacology , Dictyostelium/drug effects , Mutation/genetics , Osmotic Pressure/drug effects , Phosphorylation/drug effects , Protozoan Proteins/genetics , Pseudopodia/drug effects , Pseudopodia/metabolism , Signal Transduction/drug effects
18.
Nat Commun ; 12(1): 6157, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34697316

ABSTRACT

The pathogen Staphylococcus aureus can readily develop antibiotic resistance and evade the human immune system, which is associated with reduced levels of neutrophil recruitment. Here, we present a class of antibacterial peptides with potential to act both as antibiotics and as neutrophil chemoattractants. The compounds, which we term 'antibiotic-chemoattractants', consist of a formylated peptide (known to act as chemoattractant for neutrophil recruitment) that is covalently linked to the antibiotic vancomycin (known to bind to the bacterial cell wall). We use a combination of in vitro assays, cellular assays, infection-on-a-chip and in vivo mouse models to show that the compounds improve the recruitment, engulfment and killing of S. aureus by neutrophils. Furthermore, optimizing the formyl peptide sequence can enhance neutrophil activity through differential activation of formyl peptide receptors. Thus, we propose antibiotic-chemoattractants as an alternate approach for antibiotic development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Chemotactic Factors/pharmacology , Neutrophils/drug effects , Staphylococcus aureus/drug effects , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacterial Load/drug effects , Chemotactic Factors/chemistry , Chemotactic Factors/therapeutic use , Drug Resistance, Bacterial/drug effects , Immunotherapy , Mice , Neutrophils/immunology , Neutrophils/metabolism , Peptides/chemistry , Peptides/pharmacology , Phagocytosis/drug effects , Receptors, Formyl Peptide/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/therapy , Vancomycin/chemistry , Vancomycin/pharmacology
19.
Sci Rep ; 11(1): 16891, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34413454

ABSTRACT

Trastuzumab deruxtecan (T-DXd), a HER2-targeting antibody-drug conjugate with a topoisomerase I inhibitor deruxtecan (DXd), exhibits an excellent anti-tumor effect in previously treated HER2-positive tumors. A recent study demonstrated that T-DXd not only suppressed tumor growth but also enhanced anti-tumor immunity through increasing the number of tumor-infiltrating CD8+ T cells and enhancement of major-histocompatibility-complex class I expression on tumor cells in a mouse model. However, the effect of T-DXd on anti-tumor immune responses in human cancers is largely unknown. We investigated the effect of T-DXd on the expression of HLA class I and CXCL9/10/11, T-cell chemoattractants, in HER2-positive human gastric cancer (GC) cells. We found that T-DXd significantly inhibited GC cell proliferation in a HER2-dependent manner, while it slightly increased the expression of HLA class I in HER2-positive GC cells. Moreover, we revealed that T-DXd significantly induced mRNA expression of CXCL9/10/11 in HER2-positive GC cells. T-DXd-triggered up-regulation of these chemokines was mediated through the activation of DNA damage signaling pathways. These results suggest that T-DXd triggers anti-tumor immune responses at least in part through induction of the expression of HLA class I and CXCL9/10/11 on HER2-positive GC cells, resulting in the enhancement of anti-tumor immunity in human GC.


Subject(s)
Camptothecin/analogs & derivatives , Chemokine CXCL10/genetics , Chemokine CXCL11/genetics , Chemokine CXCL9/genetics , Histocompatibility Antigens Class I/metabolism , Immunoconjugates/therapeutic use , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/immunology , Trastuzumab/therapeutic use , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Chemotactic Factors/pharmacology , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics , Trastuzumab/pharmacology , Up-Regulation/drug effects
20.
PLoS Comput Biol ; 17(7): e1008803, 2021 07.
Article in English | MEDLINE | ID: mdl-34260581

ABSTRACT

During the last decade, a consensus has emerged that the stochastic triggering of an excitable system drives pseudopod formation and subsequent migration of amoeboid cells. The presence of chemoattractant stimuli alters the threshold for triggering this activity and can bias the direction of migration. Though noise plays an important role in these behaviors, mathematical models have typically ignored its origin and merely introduced it as an external signal into a series of reaction-diffusion equations. Here we consider a more realistic description based on a reaction-diffusion master equation formalism to implement these networks. In this scheme, noise arises naturally from a stochastic description of the various reaction and diffusion terms. Working on a three-dimensional geometry in which separate compartments are divided into a tetrahedral mesh, we implement a modular description of the system, consisting of G-protein coupled receptor signaling (GPCR), a local excitation-global inhibition mechanism (LEGI), and signal transduction excitable network (STEN). Our models implement detailed biochemical descriptions whenever this information is available, such as in the GPCR and G-protein interactions. In contrast, where the biochemical entities are less certain, such as the LEGI mechanism, we consider various possible schemes and highlight the differences between them. Our simulations show that even when the LEGI mechanism displays perfect adaptation in terms of the mean level of proteins, the variance shows a dose-dependence. This differs between the various models considered, suggesting a possible means for determining experimentally among the various potential networks. Overall, our simulations recreate temporal and spatial patterns observed experimentally in both wild-type and perturbed cells, providing further evidence for the excitable system paradigm. Moreover, because of the overall importance and ubiquity of the modules we consider, including GPCR signaling and adaptation, our results will be of interest beyond the field of directed migration.


Subject(s)
Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Computer Simulation , Models, Biological , Computational Biology , Diffusion , Pseudopodia/drug effects , Stochastic Processes
SELECTION OF CITATIONS
SEARCH DETAIL
...