Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(4): 604-611, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36879067

RESUMEN

Infection with severe acute respiratory syndrome coronavirus 2 associates with diverse symptoms, which can persist for months. While antiviral antibodies are protective, those targeting interferons and other immune factors are associated with adverse coronavirus disease 2019 (COVID-19) outcomes. Here we discovered that antibodies against specific chemokines were omnipresent post-COVID-19, were associated with favorable disease outcome and negatively correlated with the development of long COVID at 1 yr post-infection. Chemokine antibodies were also present in HIV-1 infection and autoimmune disorders, but they targeted different chemokines compared with COVID-19. Monoclonal antibodies derived from COVID-19 convalescents that bound to the chemokine N-loop impaired cell migration. Given the role of chemokines in orchestrating immune cell trafficking, naturally arising chemokine antibodies may modulate the inflammatory response and thus bear therapeutic potential.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Autoanticuerpos , Síndrome Post Agudo de COVID-19 , Quimiocinas
2.
Immunity ; 49(6): 1062-1076.e6, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30446388

RESUMEN

Neutrophils require directional cues to navigate through the complex structure of venular walls and into inflamed tissues. Here we applied confocal intravital microscopy to analyze neutrophil emigration in cytokine-stimulated mouse cremaster muscles. We identified differential and non-redundant roles for the chemokines CXCL1 and CXCL2, governed by their distinct cellular sources. CXCL1 was produced mainly by TNF-stimulated endothelial cells (ECs) and pericytes and supported luminal and sub-EC neutrophil crawling. Conversely, neutrophils were the main producers of CXCL2, and this chemokine was critical for correct breaching of endothelial junctions. This pro-migratory activity of CXCL2 depended on the atypical chemokine receptor 1 (ACKR1), which is enriched within endothelial junctions. Transmigrating neutrophils promoted a self-guided migration response through EC junctions, creating a junctional chemokine "depot" in the form of ACKR1-presented CXCL2 that enabled efficient unidirectional luminal-to-abluminal migration. Thus, CXCL1 and CXCL2 act in a sequential manner to guide neutrophils through venular walls as governed by their distinct cellular sources.


Asunto(s)
Quimiocina CXCL1 , Quimiocina CXCL2 , Sistema del Grupo Sanguíneo Duffy , Neutrófilos , Receptores de Superficie Celular , Migración Transendotelial y Transepitelial , Animales , Músculos Abdominales/efectos de los fármacos , Músculos Abdominales/inmunología , Músculos Abdominales/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/genética , Quimiocina CXCL2/inmunología , Quimiocina CXCL2/metabolismo , Sistema del Grupo Sanguíneo Duffy/genética , Sistema del Grupo Sanguíneo Duffy/inmunología , Sistema del Grupo Sanguíneo Duffy/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/inmunología , Uniones Intercelulares/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/genética , Migración Transendotelial y Transepitelial/inmunología , Factor de Necrosis Tumoral alfa/farmacología
3.
PLoS Biol ; 21(5): e3002111, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37159457

RESUMEN

Atypical chemokine receptors (ACKRs) scavenge chemokines and can contribute to gradient formation by binding, internalizing, and delivering chemokines for lysosomal degradation. ACKRs do not couple to G-proteins and fail to induce typical signaling induced by chemokine receptors. ACKR3, which binds and scavenges CXCL12 and CXCL11, is known to be expressed in vascular endothelium, where it has immediate access to circulating chemokines. ACKR4, which binds and scavenges CCL19, CCL20, CCL21, CCL22, and CCL25, has also been detected in lymphatic and blood vessels of secondary lymphoid organs, where it clears chemokines to facilitate cell migration. Recently, GPR182, a novel ACKR-like scavenger receptor, has been identified and partially deorphanized. Multiple studies point towards the potential coexpression of these 3 ACKRs, which all interact with homeostatic chemokines, in defined cellular microenvironments of several organs. However, an extensive map of ACKR3, ACKR4, and GPR182 expression in mice has been missing. In order to reliably detect ACKR expression and coexpression, in the absence of specific anti-ACKR antibodies, we generated fluorescent reporter mice, ACKR3GFP/+, ACKR4GFP/+, GPR182mCherry/+, and engineered fluorescently labeled ACKR-selective chimeric chemokines for in vivo uptake. Our study on young healthy mice revealed unique and common expression patterns of ACKRs in primary and secondary lymphoid organs, small intestine, colon, liver, and kidney. Furthermore, using chimeric chemokines, we were able to detect distinct zonal expression and activity of ACKR4 and GPR182 in the liver, which suggests their cooperative relationship. This study provides a broad comparative view and a solid stepping stone for future functional explorations of ACKRs based on the microanatomical localization and distinct and cooperative roles of these powerful chemokine scavengers.


Asunto(s)
Transducción de Señal , Animales , Ratones , Quimiocina CCL19/metabolismo , Movimiento Celular
4.
J Immunol ; 208(6): 1493-1499, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35181636

RESUMEN

Two-photon intravital microscopy (2P-IVM) has become a widely used technique to study cell-to-cell interactions in living organisms. Four-dimensional imaging data obtained via 2P-IVM are classically analyzed by performing automated cell tracking, a procedure that computes the trajectories followed by each cell. However, technical artifacts, such as brightness shifts, the presence of autofluorescent objects, and channel crosstalking, affect the specificity of imaging channels for the cells of interest, thus hampering cell detection. Recently, machine learning has been applied to overcome a variety of obstacles in biomedical imaging. However, existing methods are not tailored for the specific problems of intravital imaging of immune cells. Moreover, results are highly dependent on the quality of the annotations provided by the user. In this study, we developed CANCOL, a tool that facilitates the application of machine learning for automated tracking of immune cells in 2P-IVM. CANCOL guides the user during the annotation of specific objects that are problematic for cell tracking when not properly annotated. Then, it computes a virtual colocalization channel that is specific for the cells of interest. We validated the use of CANCOL on challenging 2P-IVM videos from murine organs, obtaining a significant improvement in the accuracy of automated tracking while reducing the time required for manual track curation.


Asunto(s)
Comunicación Celular , Microscopía Intravital , Animales , Artefactos , Rastreo Celular , Computadores , Microscopía Intravital/métodos , Ratones
5.
Immunity ; 41(5): 789-801, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25464855

RESUMEN

Microbial colonization of the gut induces the development of gut-associated lymphoid tissue (GALT). The molecular mechanisms that regulate GALT function and result in gut-commensal homeostasis are poorly defined. T follicular helper (Tfh) cells in Peyer's patches (PPs) promote high-affinity IgA responses. Here we found that the ATP-gated ionotropic P2X7 receptor controls Tfh cell numbers in PPs. Lack of P2X7 in Tfh cells enhanced germinal center reactions and high-affinity IgA secretion and binding to commensals. The ensuing depletion of mucosal bacteria resulted in reduced systemic translocation of microbial components, lowering B1 cell stimulation and serum IgM concentrations. Mice lacking P2X7 had increased susceptibility to polymicrobial sepsis, which was rescued by Tfh cell depletion or administration of purified IgM. Thus, regulation of Tfh cells by P2X7 activity is important for mucosal colonization, which in turn results in IgM serum concentrations necessary to protect the host from bacteremia.


Asunto(s)
Mucosa Intestinal/inmunología , Ganglios Linfáticos Agregados/inmunología , Receptores Purinérgicos P2X7/inmunología , Simbiosis/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Adenosina Trifosfato/metabolismo , Animales , Linfocitos B/inmunología , Bacteriemia/inmunología , Predisposición Genética a la Enfermedad , Centro Germinal/inmunología , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina M/sangre , Mucosa Intestinal/microbiología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/inmunología , Ganglios Linfáticos Agregados/citología , Receptores Purinérgicos P2X7/genética , Sepsis/inmunología , Sepsis/microbiología
8.
Nat Immunol ; 9(9): 953-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18711432

RESUMEN

A prominent activity of the chemokine system is the regulation of leukocyte trafficking. Here we summarize recent findings on the initial steps in chemokine receptor-induced signal transduction in leukocytes. In particular, we discuss the potential influences of the formation of oligomers of ligand and receptor and of coupling between chemokine signals and regulators of the cytoskeleton, such as small GTPases.


Asunto(s)
Quimiocinas/inmunología , Quimiocinas/metabolismo , Leucocitos/inmunología , Transducción de Señal , Animales , GTP Fosfohidrolasas/metabolismo , Humanos , Activación de Linfocitos , Receptores de Quimiocina/metabolismo
9.
Cytokine ; 109: 72-75, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29433906

RESUMEN

Migration of leukocytes is typically mediated by G protein-coupled receptors (GPCRs) upon activation by specific ligands that range from small peptides, chemokines to a variety of lipidic molecules. The heptahelical receptors are highly dynamic structures and the signaling efficiency largely depends on the discrete contact with the ligand. In addition, several allosteric modulators of receptor activity have been reported, which do not induce migration by themselves. Another important mechanism modulating the activity of GPCRs is their local environment. Not only the membrane lipid composition influences the activity, but also direct binding of lipids, in particular cholesterol, was shown to alter receptor signaling properties. Recent findings indicate that also chemokine receptor activity is modulated by membrane lipids. In this short review we discuss this new paradigm and potential consequences for chemokine-induced migration.


Asunto(s)
Movimiento Celular/fisiología , Quimiocinas/metabolismo , Colesterol/metabolismo , Leucocitos/metabolismo , Lípidos de la Membrana/metabolismo , Receptores de Quimiocina/metabolismo , Membrana Celular/metabolismo , Humanos , Transducción de Señal/fisiología
10.
FASEB J ; 31(7): 3084-3097, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28360196

RESUMEN

The chemokine receptor, CXC chemokine receptor 4 (CXCR4), is selective for CXC chemokine ligand 12 (CXCL12), is broadly expressed in blood and tissue cells, and is essential during embryogenesis and hematopoiesis. CXCL14 is a homeostatic chemokine with unknown receptor selectivity and preferential expression in peripheral tissues. Here, we demonstrate that CXCL14 synergized with CXCL12 in the induction of chemokine responses in primary human lymphoid cells and cell lines that express CXCR4. Combining subactive concentrations of CXCL12 with 100-300 nM CXCL14 resulted in chemotaxis responses that exceeded maximal responses that were obtained with CXCL12 alone. CXCL14 did not activate CXCR4-expressing cells (i.e., failed to trigger chemotaxis and Ca2+ mobilization, as well as signaling via ERK1/2 and the small GTPase Rac1); however, CXCL14 bound to CXCR4 with high affinity, induced redistribution of cell-surface CXCR4, and enhanced HIV-1 infection by >3-fold. We postulate that CXCL14 is a positive allosteric modulator of CXCR4 that enhances the potency of CXCR4 ligands. Our findings provide new insights that will inform the development of novel therapeutics that target CXCR4 in a range of diseases, including cancer, autoimmunity, and HIV.-Collins, P. J., McCully, M. L., Martínez-Muñoz, L., Santiago, C., Wheeldon, J., Caucheteux, S., Thelen, S., Cecchinato, V., Laufer, J. M., Purvanov, V., Monneau, Y. R., Lortat-Jacob, H., Legler, D. F., Uguccioni, M., Thelen, M., Piguet, V., Mellado, M., Moser, B. Epithelial chemokine CXCL14 synergizes with CXCL12 via allosteric modulation of CXCR4.


Asunto(s)
Quimiocina CXCL12/metabolismo , Quimiocinas CXC/metabolismo , Regulación de la Expresión Génica/fisiología , Leucocitos Mononucleares/metabolismo , Receptores CXCR4/metabolismo , Secuencia de Aminoácidos , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocinas CXC/genética , Quimiotaxis , VIH-1/fisiología , Humanos , Unión Proteica , Conformación Proteica , ARN Mensajero , Receptores CXCR4/genética , Transducción de Señal
11.
Mol Pharmacol ; 91(4): 331-338, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28082305

RESUMEN

Chemokine receptors are seven transmembrane-domain receptors belonging to class A of G-protein-coupled receptors (GPCRs). The receptors together with their chemokine ligands constitute the chemokine system, which is essential for directing cell migration and plays a crucial role in a variety of physiologic and pathologic processes. Given the importance of orchestrating cell migration, it is vital that chemokine receptor signaling is tightly regulated to ensure appropriate responses. Recent studies highlight a key role for cholesterol in modulating chemokine receptor activities. The steroid influences the spatial organization of GPCRs within the membrane bilayer, and consequently can tune chemokine receptor signaling. The effects of cholesterol on the organization and function of chemokine receptors and GPCRs in general include direct and indirect effects (Fig. 1). Here, we review how cholesterol and some key metabolites modulate functions of the chemokine system in multiple ways. We emphasize the role of cholesterol in chemokine receptor oligomerization, thereby promoting the formation of a signaling hub enabling integration of distinct signaling pathways at the receptor-membrane interface. Moreover, we discuss the role of cholesterol in stabilizing particular receptor conformations and its consequence for chemokine binding. Finally, we highlight how cholesterol accumulation, its deprivation, or cholesterol metabolites contribute to modulating cell orchestration during inflammation, induction of an adaptive immune response, as well as to dampening an anti-tumor immune response.


Asunto(s)
Colesterol/metabolismo , Receptores de Quimiocina/metabolismo , Animales , Membrana Celular/metabolismo , Enfermedad , Humanos , Modelos Biológicos , Receptores de Quimiocina/química , Transducción de Señal
12.
Development ; 141(9): 1857-63, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24718993

RESUMEN

The CXCL12/CXCR4 signaling pathway is involved in the development of numerous neuronal and non-neuronal structures. Recent work established that the atypical second CXCL12 receptor, CXCR7, is essential for the proper migration of interneuron precursors in the developing cerebral cortex. Two CXCR7-mediated functions were proposed in this process: direct modulation of ß-arrestin-mediated signaling cascades and CXCL12 scavenging to regulate local chemokine availability and ensure responsiveness of the CXCL12/CXCR4 pathway in interneurons. Neither of these functions has been proven in the embryonic brain. Here, we demonstrate that migrating interneurons efficiently sequester CXCL12 through CXCR7. CXCR7 ablation causes excessive phosphorylation and downregulation of CXCR4 throughout the cortex in mice expressing CXCL12, but not in CXCL12-deficient animals. Cxcl12(-/-) mice lack activated CXCR4 in embryonic brain lysates and display a similar interneuron positioning defect as Cxcr4(-/-), Cxcr7(-/-) and Cxcl12(-/-);Cxcr7(-/-) animals. Thus, CXCL12 is the only CXCR4-activating ligand in the embryonic brain and deletion of one of the CXCL12 receptors is sufficient to generate a migration phenotype that corresponds to the CXCL12-deficient pathway. Our findings imply that interfering with the CXCL12-scavenging activity of CXCR7 causes loss of CXCR4 function as a consequence of excessive CXCL12-mediated CXCR4 activation and degradation.


Asunto(s)
Movimiento Celular , Corteza Cerebral/citología , Quimiocina CXCL12/metabolismo , Regulación hacia Abajo , Interneuronas/citología , Interneuronas/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Animales , Embrión de Mamíferos/citología , Células HEK293 , Humanos , Ratones , Modelos Biológicos , Proteínas Recombinantes de Fusión/metabolismo
13.
Pharmacol Rev ; 66(1): 1-79, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24218476

RESUMEN

Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.


Asunto(s)
Receptores de Quimiocina , Animales , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/metabolismo , Humanos , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Receptores de Quimiocina/clasificación , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Terminología como Asunto , Garrapatas , Proteínas Virales/genética , Proteínas Virales/metabolismo
14.
Chimia (Aarau) ; 70(12): 856-859, 2016 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-28661356

RESUMEN

The in vitro synthesis of correctly folded functional proteins remains challenging. Chemokines, which consist of only 70-100 amino acids, are accessible through solid-phase synthesis and easily fold into a thermally stable tertiary structure. From the time of their discovery in the late 1980s chemokines could therefore be synthesized using biochemical and chemical protocols for structure-function analyses and for exploring the chemokine system in vitro and in vivo. In this short overview aimed at a chemistry-oriented readership we will introduce chemokines in general, and then discuss their structure, their isolation from biological materials, as well as the different methods to produce chemokines in the laboratory and finally we will present some examples of their functions in vivo.


Asunto(s)
Quimiocinas/química , Quimiocinas/metabolismo , Quimiocinas/síntesis química , Quimiocinas/genética , Humanos
15.
Eur J Immunol ; 44(3): 694-705, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24259140

RESUMEN

The atypical chemokine receptor CXCR7 binds the chemokines CXCL12 and CXCL11. The receptor is widely expressed and was shown to tune CXCR12-induced responses of CXCR4. Here, the function of CXCR7 was examined at late stages of human B-cell maturation, when B cells differentiate into Ab-secreting plasmablasts. We identified two populations of CXCR7(+) cells in tonsillar lymphocytes, one being presumably memory B cells or early plasmablasts (FSC(low) CD19(+) CD38(mid) ) and the other being plasmablasts or early plasma cells (FSC(high) CD19(+) CD38(+) ). CXCR7 is expressed on CD19(+) CD27(+) memory B cells, on CD19(+) CD38(+) CD138(-) and intracellular immunoglobulin high plasmablasts, but not on CD19(+) CD138(+) icIg(high) plasma cells. The differential expression pattern suggests a potential contribution of the scavenger receptor in final B-cell maturation. On in vitro differentiating B cells, we found a marked inverse correlation between CXCR7 and CXCR5 cell surface levels, whereas expression of CXCR4 remained almost constant. Migration assays performed with tonsillar mononuclear cells or in vitro differentiated cells revealed that inhibition of CXCR7 markedly increases chemotaxis toward CXCL12, especially at late stages of B-cell maturation. Chemotaxis was attenuated in the presence of CXCR4 antagonists, confirming that migration is CXCR4 mediated. Our findings unequivocally demonstrate a novel role for CXCR7 in regulating the migration of plasmablasts during B-cell maturation.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Movimiento Celular/fisiología , Receptores CXCR/metabolismo , Linfocitos B/citología , Diferenciación Celular , Regulación de la Expresión Génica , Humanos , Memoria Inmunológica , Inmunofenotipificación , Leucocitos Mononucleares , Modelos Inmunológicos , Tonsila Palatina/citología , Tonsila Palatina/metabolismo , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores CXCR/genética , Receptores CXCR4/metabolismo , Receptores CXCR5/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
16.
Development ; 138(14): 2909-14, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21693511

RESUMEN

The active migration of primordial germ cells (PGCs) from their site of specification towards their target is a valuable model for investigating directed cell migration within the complex environment of the developing embryo. In several vertebrates, PGC migration is guided by Cxcl12, a member of the chemokine superfamily. Interestingly, two distinct Cxcl12 paralogs are expressed in zebrafish embryos and contribute to the chemotattractive landscape. Although this offers versatility in the use of chemokine signals, it also requires a mechanism through which migrating cells prioritize the relevant cues that they encounter. Here, we show that PGCs respond preferentially to one of the paralogs and define the molecular basis for this biased behavior. We find that a single amino acid exchange switches the relative affinity of the Cxcl12 ligands for one of the duplicated Cxcr4 receptors, thereby determining the functional specialization of each chemokine that elicits a distinct function in a distinct process. This scenario represents an example of protein subfunctionalization--the specialization of two gene copies to perform complementary functions following gene duplication--which in this case is based on receptor-ligand interaction. Such specialization increases the complexity and flexibility of chemokine signaling in controlling concurrent developmental processes.


Asunto(s)
Movimiento Celular/fisiología , Quimiocina CXCL12/metabolismo , Evolución Molecular , Células Germinativas/fisiología , Receptores CXCR4/metabolismo , Pez Cebra/embriología , Sustitución de Aminoácidos , Animales , Línea Celular , Quimiocina CXCL12/genética , Técnicas de Silenciamiento del Gen , Humanos , Hibridación in Situ , Microscopía Confocal , Espectrometría de Fluorescencia , Pez Cebra/metabolismo
17.
PLoS Pathog ; 8(2): e1002497, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22319442

RESUMEN

Herpes simplex virus (HSV) types 1 and 2 are highly prevalent human neurotropic pathogens that cause a variety of diseases, including lethal encephalitis. The relationship between HSV and the host immune system is one of the main determinants of the infection outcome. Chemokines play relevant roles in antiviral response and immunopathology, but the modulation of chemokine function by HSV is not well understood. We have addressed the modulation of chemokine function mediated by HSV. By using surface plasmon resonance and crosslinking assays we show that secreted glycoprotein G (SgG) from both HSV-1 and HSV-2 binds chemokines with high affinity. Chemokine binding activity was also observed in the supernatant of HSV-2 infected cells and in the plasma membrane of cells infected with HSV-1 wild type but not with a gG deficient HSV-1 mutant. Cell-binding and competition experiments indicate that the interaction takes place through the glycosaminoglycan-binding domain of the chemokine. The functional relevance of the interaction was determined both in vitro, by performing transwell assays, time-lapse microscopy, and signal transduction experiments; and in vivo, using the air pouch model of inflammation. Interestingly, and in contrast to what has been observed for previously described viral chemokine binding proteins, HSV SgGs do not inhibit chemokine function. On the contrary, HSV SgGs enhance chemotaxis both in vitro and in vivo through increasing directionality, potency and receptor signaling. This is the first report, to our knowledge, of a viral chemokine binding protein from a human pathogen that increases chemokine function and points towards a previously undescribed strategy of immune modulation mediated by viruses.


Asunto(s)
Quimiocinas/metabolismo , Herpes Simple/patología , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 2/metabolismo , Interacciones Huésped-Patógeno , Proteínas del Envoltorio Viral/metabolismo , Animales , Células Cultivadas , Quimiotaxis , Femenino , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Herpesvirus Humano 2/inmunología , Herpesvirus Humano 2/patogenicidad , Factores Inmunológicos/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología
18.
Front Immunol ; 15: 1358800, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38803493

RESUMEN

During inflammation and tissue regeneration, the alarmin High Mobility Group Box 1 (HMGB1), in its reduced isoform, enhances the activity of the chemokine CXCL12, forming a heterocomplex that acts via the chemokine receptor CXCR4. Despite the established roles of both HMGB1 and CXCL12 in tumor progression and metastatic spread to distal sites, the role of the CXCL12/HMGB1 heterocomplex in cancer has never been investigated. By employing a newly established mass spectrometry protocol that allows an unambiguous distinction between reduced (red-HMGB1) and oxidized (ox-HMGB1) HMGB1 isoforms in cell lysates, we demonstrate that human epithelial cells derived from breast (MCF-7 and MDA-MB-231) and prostate (PC-3) cancer predominantly express red-HMGB1, while primary CD3+ T lymphocytes from peripheral blood express both HMGB1 isoforms. All these cancer cells release HMGB1 in the extracellular microenvironment together with varying concentrations of thioredoxin and thioredoxin reductase. The CXCL12/HMGB1 heterocomplex enhances, via CXCR4, the directional migration and invasiveness of cancer cells characterized by high metastatic potential that possess a fully active thioredoxin system, contributing to maintain red-HMGB1. On the contrary, cancer cells with low metastatic potential, lack thioredoxin reductase, promptly uptake CXCL12 and fail to respond to the heterocomplex. Our study demonstrates that the responsiveness of cancer cells to the CXCL12/HMGB1 heterocomplex, resulting in enhanced cell migration and invasiveness, depends on the maintenance of HMGB1 in its reduced isoform, and suggests disruption of the heterocomplex as a potential therapeutic target to inhibit invasion and metastatic spread in cancer therapies.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12 , Proteína HMGB1 , Isoformas de Proteínas , Receptores CXCR4 , Humanos , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Quimiocina CXCL12/metabolismo , Línea Celular Tumoral , Femenino , Microambiente Tumoral , Masculino , Neoplasias/metabolismo , Neoplasias/patología
19.
Elife ; 122024 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-38497754

RESUMEN

Intravital microscopy has revolutionized live-cell imaging by allowing the study of spatial-temporal cell dynamics in living animals. However, the complexity of the data generated by this technology has limited the development of effective computational tools to identify and quantify cell processes. Amongst them, apoptosis is a crucial form of regulated cell death involved in tissue homeostasis and host defense. Live-cell imaging enabled the study of apoptosis at the cellular level, enhancing our understanding of its spatial-temporal regulation. However, at present, no computational method can deliver robust detection of apoptosis in microscopy timelapses. To overcome this limitation, we developed ADeS, a deep learning-based apoptosis detection system that employs the principle of activity recognition. We trained ADeS on extensive datasets containing more than 10,000 apoptotic instances collected both in vitro and in vivo, achieving a classification accuracy above 98% and outperforming state-of-the-art solutions. ADeS is the first method capable of detecting the location and duration of multiple apoptotic events in full microscopy timelapses, surpassing human performance in the same task. We demonstrated the effectiveness and robustness of ADeS across various imaging modalities, cell types, and staining techniques. Finally, we employed ADeS to quantify cell survival in vitro and tissue damage in mice, demonstrating its potential application in toxicity assays, treatment evaluation, and inflammatory dynamics. Our findings suggest that ADeS is a valuable tool for the accurate detection and quantification of apoptosis in live-cell imaging and, in particular, intravital microscopy data, providing insights into the complex spatial-temporal regulation of this process.


Asunto(s)
Apoptosis , Microscopía , Humanos , Animales , Ratones , Supervivencia Celular , Microscopía Intravital , Reconocimiento en Psicología
20.
Biochem J ; 443(1): 173-83, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22242915

RESUMEN

P-Rex1 is a GEF (guanine-nucleotide-exchange factor) for the small G-protein Rac that is activated by PIP3 (phosphatidylinositol 3,4,5-trisphosphate) and Gßγ subunits and inhibited by PKA (protein kinase A). In the present study we show that PP1α (protein phosphatase 1α) binds P-Rex1 through an RVxF-type docking motif. PP1α activates P-Rex1 directly in vitro, both independently of and additively to PIP3 and Gßγ. PP1α also substantially activates P-Rex1 in vivo, both in basal and PDGF (platelet-derived growth factor)- or LPA (lysophosphatidic acid)-stimulated cells. The phosphatase activity of PP1α is required for P-Rex1 activation. PP1ß, a close homologue of PP1α, is also able to activate P-Rex1, but less effectively. PP1α stimulates P-Rex1-mediated Rac-dependent changes in endothelial cell morphology. MS analysis of wild-type P-Rex1 and a PP1α-binding-deficient mutant revealed that endogenous PP1α dephosphorylates P-Rex1 on at least three residues, Ser834, Ser1001 and Ser1165. Site-directed mutagenesis of Ser1165 to alanine caused activation of P-Rex1 to a similar degree as did PP1α, confirming Ser1165 as a dephosphorylation site important in regulating P-Rex1 Rac-GEF activity. In summary, we have identified a novel mechanism for direct activation of P-Rex1 through PP1α-dependent dephosphorylation.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/química , Proteína Fosfatasa 1/química , Secuencias de Aminoácidos , Animales , Aorta/citología , Forma de la Célula , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Unión Proteica , Proteína Fosfatasa 1/metabolismo , Estructura Terciaria de Proteína , Conejos , Porcinos , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA