Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(21): e2119483119, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35588454

RESUMEN

Chemokine receptor nanoscale organization at the cell membrane is orchestrated by the actin cytoskeleton and influences cell responses. Using single-particle tracking analysis we show that CXCR4R334X, a truncated mutant chemokine receptor linked to WHIM syndrome (warts, hypogammaglobulinemia, infections, myelokathexis), fails to nanoclusterize after CXCL12 stimulation, and alters the lateral mobility and spatial organization of CXCR4 when coexpressed. These findings correlate with multiple phalloidin-positive protrusions in cells expressing CXCR4R334X, and their inability to correctly sense chemokine gradients. The underlying mechanisms involve inappropriate actin cytoskeleton remodeling due to the inadequate ß-arrestin1 activation by CXCR4R334X, which disrupts the equilibrium between activated and deactivated cofilin. Overall, we provide insights into the molecular mechanisms governing CXCR4 nanoclustering, signaling and cell function, and highlight the essential scaffold role of ß-arrestin1 to support CXCL12-mediated actin reorganization and receptor clustering. These defects associated with CXCR4R334X expression might contribute to the severe immunological symptoms associated with WHIM syndrome.


Asunto(s)
Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4 , Verrugas , Factores Despolimerizantes de la Actina/metabolismo , Membrana Celular/metabolismo , Movimiento Celular , Humanos , Mutación , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Imagen Individual de Molécula , Verrugas/genética , Verrugas/metabolismo
2.
Biochem Biophys Res Commun ; 528(2): 347-358, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32145914

RESUMEN

The chemokines receptor family are membrane-expressed class A-specific seven-transmembrane receptors linked to G proteins. Through interaction with the corresponding ligands, the chemokines, they induce a wide variety of cellular responses including cell polarization, movement, immune and inflammatory responses, as well as the prevention of HIV-1 infection. Like a Russian matryoshka doll, the chemokine receptor system is more complex than initially envisaged. This review focuses on the mechanisms that contribute to this dazzling complexity and how they modulate the signaling events triggered by chemokines. The chemokines and their receptors exist as monomers, dimers and oligomers, their expression pattern is highly regulated, and the ligands can bind distinct receptors with similar affinities. The use of novel imaging-based technologies, particularly real-time imaging modalities, has shed new light on the very dynamic conformations that chemokine receptors adopt depending on the cellular context, and that affect chemokine-mediated responses. This complex scenario presents both challenging and exciting opportunities for drug discovery.


Asunto(s)
Receptores de Quimiocina/metabolismo , Animales , Quimiocinas/química , Quimiocinas/metabolismo , Factores Quimiotácticos/metabolismo , Humanos , Multimerización de Proteína
3.
J Immunol ; 198(1): 184-195, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27895171

RESUMEN

CD4+ T cell repopulation of the gut is rarely achieved in HIV-1-infected individuals who are receiving clinically effective antiretroviral therapy. Alterations in the integrity of the mucosal barrier have been indicated as a cause for chronic immune activation and disease progression. In this study, we present evidence that persistent immune activation causes impairment of lymphocytes to respond to chemotactic stimuli, thus preventing their trafficking from the blood stream to peripheral organs. CCR6+ and CXCR3+ Th cells accumulate in the blood of aviremic HIV-1-infected patients on long-term antiretroviral therapy, and their frequency in the circulation positively correlates to levels of soluble CD14 in plasma, a marker of chronic immune activation. Th cells show an impaired response to chemotactic stimuli both in humans and in the pathogenic model of SIV infection, and this defect is due to hyperactivation of cofilin and inefficient actin polymerization. Taking advantage of a murine model of chronic immune activation, we demonstrate that cytoskeleton remodeling, induced by okadaic acid, restores lymphocyte migration in response to chemokines, both in vitro and in vivo. This study calls for novel pharmacological approaches in those pathological conditions characterized by persistent immune activation and loss of trafficking of T cell subsets to niches that sustain their maturation and activities.


Asunto(s)
Actinas/metabolismo , Quimiotaxis de Leucocito/inmunología , Infecciones por VIH/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Separación Celular , Citoesqueleto/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , VIH-1 , Humanos , Inmunohistoquímica , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Polimerizacion , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CCR6/inmunología , Receptores CXCR3/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología
4.
J Neurochem ; 136(6): 1219-1231, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26710242

RESUMEN

Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders, characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta. PD mostly occurs sporadically and its cause remains unknown, nevertheless the discovery of familiar forms of PD, characterized by mutations of genes encoding proteins associated with mitochondria homeostasis, suggests a strong implication of the mitochondrial quality control system in PD. We investigated the effect of dopamine cytosolic accumulation in undifferentiated SH-SY5Y cells, an in vitro model widely used to reproduce impairment of dopamine homeostasis, an early step in PD pathogenesis. A strong depolarization of the mitochondrial membrane was observed after dopamine exposure. Nevertheless, mitochondrial network resulted to assume a peculiar morphology with a distinct pattern of OPA1 and MFN1, key regulators of mitochondrial dynamics. Moreover, selective elimination of dysfunctional mitochondria did not take place, suggesting an impairment of the mitophagic machinery induced by dopamine. Indeed, PINK1 did not accumulate on the outer mitochondrial membrane, nor was parkin recruited to depolarized mitochondria. Altogether, our results indicate that an improper handling of dysfunctional mitochondria may be a leading event in PD pathogenesis. Impaired dopamine (DA) homeostasis and oxidative stress play a key role in the pathogenesis of Parkinson's disease. Free cytosolic dopamine undergoes spontaneous oxidation and generates semiquinonic and quinonic species (DAQ) with the concurrent production of reactive oxygen species (ROS). Dopamine dissipates mitochondrial potential (Δψm ) with a peculiar alteration of the mitochondrial network. However, PINK1-dependent mitophagy is not activated by dopamine toxicity and dysfunctional mitochondria accumulate inside the cell.

5.
Biochim Biophys Acta ; 1842(9): 1816-22, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24998333

RESUMEN

Altered dopamine homeostasis plays a key role in the pathogenesis of Parkinson's disease. The generation of reactive oxygen species by spontaneous dopamine oxidation impairs mitochondrial function, causing in turn an enhancement of oxidative stress. Recent findings have highlighted the role of mitochondrial outer membrane proteins in the regulation of the correct disposal of damaged mitochondria. Here, we report the effect of altered dopamine homeostasis on the mitochondrial functionality in human neuroblastoma SH-SY5Y cells, a cellular model widely used to reproduce impaired dopamine homeostasis. We observed that dopamine significantly and relevantly reduces VDAC1 and VDAC2 levels without any change in the mRNA levels. Although mitochondria are depolarized by dopamine and mitochondrial calcium influx is reduced, dysfunctional mitochondria are not removed by mitophagy as it would be expected. Thus, alteration of dopamine homeostasis induces a mitochondrial depolarization not counteracted by the mitophagy quality control. As a consequence, the elimination of VDACs may contribute to the altered mitochondrial disposal in PD pathogenesis, thus enhancing the role of oxidative stress.


Asunto(s)
Dopamina/metabolismo , Homeostasis , Mitocondrias/patología , Neuroblastoma/patología , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Canal Aniónico 2 Dependiente del Voltaje/metabolismo , Western Blotting , Calcio/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Oxidación-Reducción , ARN Mensajero/genética , Especies Reactivas de Oxígeno , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Canal Aniónico 1 Dependiente del Voltaje/genética , Canal Aniónico 2 Dependiente del Voltaje/genética
6.
Elife ; 122024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38597186

RESUMEN

Epithelial intercellular adhesion molecule (ICAM)-1 is apically polarized, interacts with, and guides leukocytes across epithelial barriers. Polarized hepatic epithelia organize their apical membrane domain into bile canaliculi and ducts, which are not accessible to circulating immune cells but that nevertheless confine most of ICAM-1. Here, by analyzing ICAM-1_KO human hepatic cells, liver organoids from ICAM-1_KO mice and rescue-of-function experiments, we show that ICAM-1 regulates epithelial apicobasal polarity in a leukocyte adhesion-independent manner. ICAM-1 signals to an actomyosin network at the base of canalicular microvilli, thereby controlling the dynamics and size of bile canalicular-like structures. We identified the scaffolding protein EBP50/NHERF1/SLC9A3R1, which connects membrane proteins with the underlying actin cytoskeleton, in the proximity interactome of ICAM-1. EBP50 and ICAM-1 form nano-scale domains that overlap in microvilli, from which ICAM-1 regulates EBP50 nano-organization. Indeed, EBP50 expression is required for ICAM-1-mediated control of BC morphogenesis and actomyosin. Our findings indicate that ICAM-1 regulates the dynamics of epithelial apical membrane domains beyond its role as a heterotypic cell-cell adhesion molecule and reveal potential therapeutic strategies for preserving epithelial architecture during inflammatory stress.


Asunto(s)
Actomiosina , Molécula 1 de Adhesión Intercelular , Animales , Ratones , Humanos , Actomiosina/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Células Epiteliales/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Citoesqueleto de Actina/metabolismo , Leucocitos/metabolismo , Polaridad Celular
7.
Artículo en Inglés | MEDLINE | ID: mdl-35627487

RESUMEN

(1) Background: During the COVID-19 outbreak in the Lazio region, a surge in emergency medical service (EMS) calls has been observed. The objective of present study is to investigate if there is any correlation between the variation in numbers of daily EMS calls, and the short-term evolution of the epidemic wave. (2) Methods: Data from the COVID-19 outbreak has been retrieved in order to draw the epidemic curve in the Lazio region. Data from EMS calls has been used in order to determine Excess of Calls (ExCa) in the 2020−2021 years, compared to the year 2019 (baseline). Multiple linear regression models have been run between ExCa and the first-order derivative (D') of the epidemic wave in time, each regression model anticipating the epidemic progression (up to 14 days), in order to probe a correlation between the variables. (3) Results: EMS calls variation from baseline is correlated with the slope of the curve of ICU admissions, with the most fitting value found at 7 days (R2 0.33, p < 0.001). (4) Conclusions: EMS calls deviation from baseline allows public health services to predict short-term epidemic trends in COVID-19 outbreaks, and can be used as validation of current data, or as an independent estimator of future trends.


Asunto(s)
COVID-19 , Servicios Médicos de Urgencia , Epidemias , COVID-19/epidemiología , Brotes de Enfermedades , Humanos , Análisis de Series de Tiempo Interrumpido
8.
Front Immunol ; 13: 925559, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35903108

RESUMEN

Sphingolipids, ceramides and cholesterol are integral components of cellular membranes, and they also play important roles in signal transduction by regulating the dynamics of membrane receptors through their effects on membrane fluidity. Here, we combined biochemical and functional assays with single-particle tracking analysis of diffusion in the plasma membrane to demonstrate that the local lipid environment regulates CXCR4 organization and function and modulates chemokine-triggered directed cell migration. Prolonged treatment of T cells with bacterial sphingomyelinase promoted the complete and sustained breakdown of sphingomyelins and the accumulation of the corresponding ceramides, which altered both membrane fluidity and CXCR4 nanoclustering and dynamics. Under these conditions CXCR4 retained some CXCL12-mediated signaling activity but failed to promote efficient directed cell migration. Our data underscore a critical role for the local lipid composition at the cell membrane in regulating the lateral mobility of chemokine receptors, and their ability to dynamically increase receptor density at the leading edge to promote efficient cell migration.


Asunto(s)
Receptores CXCR4 , Esfingomielinas , Movimiento Celular , Ceramidas/metabolismo , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/metabolismo , Humanos , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Transducción de Señal , Linfocitos T/metabolismo
9.
J Med Chem ; 64(18): 13439-13450, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34510899

RESUMEN

During inflammatory reactions, the production and release of chemotactic factors guide the recruitment of selective leukocyte subpopulations. The alarmin HMGB1 and the chemokine CXCL12, both released in the microenvironment, can form a heterocomplex, which exclusively acts on the chemokine receptor CXCR4, enhancing cell migration, and in some pathological conditions such as rheumatoid arthritis exacerbates the immune response. An excessive cell influx at the inflammatory site can be diminished by disrupting the heterocomplex. Here, we report the computationally driven identification of the first peptide (HBP08) binding HMGB1 and selectively inhibiting the activity of the CXCL12/HMGB1 heterocomplex. Furthermore, HBP08 binds HMGB1 with the highest affinity reported so far (Kd of 0.8 ± 0.4 µM). The identification of this peptide represents an important step toward the development of innovative pharmacological tools for the treatment of severe chronic inflammatory conditions characterized by an uncontrolled immune response.


Asunto(s)
Quimiocina CXCL12/antagonistas & inhibidores , Proteína HMGB1/antagonistas & inhibidores , Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Proteína HMGB1/metabolismo , Humanos , Ratones , Simulación del Acoplamiento Molecular , Péptidos/metabolismo , Receptores CXCR4/metabolismo
10.
Front Immunol ; 11: 720, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32391018

RESUMEN

Chemokines are essential for guiding cell migration. Atypical chemokine receptors (ACKRs) contribute to the cell migration process by binding, internalizing and degrading local chemokines, which enables the formation of confined gradients. ACKRs are heptahelical membrane spanning molecules structurally related to G-protein coupled receptors (GPCRs), but seem to be unable to signal through G-proteins upon ligand binding. ACKR4 internalizes the chemokines CCL19, CCL21, and CCL25 and is best known for shaping functional CCL21 gradients. Ligand binding to ACKR4 has been shown to recruit ß-arrestins that has led to the assumption that chemokine scavenging relies on ß-arrestin-mediated ACKR4 trafficking, a common internalization route taken by class A GPCRs. Here, we show that CCL19, CCL21, and CCL25 readily recruited ß-arrestin1 and ß-arrestin2 to human ACKR4, but found no evidence for ß-arrestin-dependent or independent ACKR4-mediated activation of the kinases Erk1/2, Akt, or Src. However, we demonstrate that ß-arrestins interacted with ACKR4 in the steady-state and contributed to the spontaneous trafficking of the receptor in the absence of chemokines. Deleting the C-terminus of ACKR4 not only interfered with the interaction of ß-arrestins, but also with the uptake of fluorescently labeled cognate chemokines. We identify the GPCR kinase GRK3, and to a lesser extent GRK2, but not GRK4, GRK5, and GRK6, to be recruited to chemokine-stimulated ACKR4. We show that GRK3 recruitment proceded the recruitment of ß-arrestins upon ACKR4 engagement and that GRK2/3 inhibition partially interfered with steady-state interaction and chemokine-driven recruitment of ß-arrestins to ACKR4. Overexpressing ß-arrestin2 accelerated the uptake of fluorescently labeled CCL19, indicating that ß-arrestins contribute to the chemokine scavenging activity of ACKR4. By contrast, cells lacking ß-arrestins were still capable to take up fluorescently labeled CCL19 demonstrating that ß-arrestins are dispensable for chemokine scavenging by ACKR4.


Asunto(s)
Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Quimiocinas CC/metabolismo , Quinasa 3 del Receptor Acoplado a Proteína-G/metabolismo , Receptores CCR/metabolismo , Transducción de Señal/genética , beta-Arrestina 1/metabolismo , Arrestina beta 2/metabolismo , Células HeLa , Humanos , Plásmidos/genética , Plásmidos/metabolismo , Unión Proteica/genética , Receptores CCR/genética , Receptores CCR7/genética , Receptores CCR7/metabolismo , Transfección , Arrestina beta 2/genética
11.
Front Immunol ; 11: 550824, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33072091

RESUMEN

The chemokine receptor CXCR4 plays a fundamental role in homeostasis and pathology by orchestrating recruitment and positioning of immune cells, under the guidance of a CXCL12 gradient. The ability of chemokines to form heterocomplexes, enhancing their function, represents an additional level of regulation on their cognate receptors. In particular, the multi-faceted activity of the heterocomplex formed between CXCL12 and the alarmin HMGB1 is emerging as an unexpected player able to modulate a variety of cell responses, spanning from tissue regeneration to chronic inflammation. Nowadays, little is known on the selective signaling pathways activated when CXCR4 is triggered by the CXCL12/HMGB1 heterocomplex. In the present work, we demonstrate that this heterocomplex acts as a CXCR4 balanced agonist, activating both G protein and ß-arrestins-mediated signaling pathways to sustain chemotaxis. We generated ß-arrestins knock out HeLa cells by CRISPR/Cas9 technology and show that the CXCL12/HMGB1 heterocomplex-mediated actin polymerization is primarily ß-arrestin1 dependent, while chemotaxis requires both ß-arrestin1 and ß-arrestin2. Triggering of CXCR4 with the CXCL12/HMGB1 heterocomplex leads to an unexpected receptor retention on the cell surface, which depends on ß-arrestin2. In conclusion, the CXCL12/HMGB1 heterocomplex engages the ß-arrestin proteins differently from CXCL12, promoting a prompt availability of CXCR4 on the cell surface, and enhancing directional cell migration. These data unveil the signaling induced by the CXCL12/HMGB1 heterocomplex in view of identifying biased CXCR4 antagonists or agonists targeting the variety of functions it exerts.


Asunto(s)
Quimiocina CXCL12/metabolismo , Proteína HMGB1/metabolismo , Receptores CXCR4/metabolismo , beta-Arrestina 1/metabolismo , Arrestina beta 2/metabolismo , Actinas/química , Actinas/metabolismo , Sistemas CRISPR-Cas , Quimiotaxis , Edición Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Complejos Multiproteicos/metabolismo , Unión Proteica , Multimerización de Proteína , Transporte de Proteínas , beta-Arrestina 1/genética , Arrestina beta 2/genética
12.
Comput Struct Biotechnol J ; 17: 886-894, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31333815

RESUMEN

High-mobility Group Box 1 (HMGB1) is an abundant protein present in all mammalian cells and involved in several processes. During inflammation or tissue damage, HMGB1 is released in the extracellular space and, depending on its redox state, can form a heterocomplex with CXCL12. The heterocomplex acts exclusively via the chemokine receptor CXCR4 enhancing leukocyte recruitment. Here, we used multi-microsecond molecular dynamics (MD) simulations to elucidate the effect of the disulfide bond on the structure and dynamics of HMGB1. The results of the MD simulations show that the presence or lack of the disulfide bond between Cys23 and Cys45 modulates the conformational space explored by HMGB1, making the reduced protein more suitable to form a complex with CXCL12.

13.
Front Immunol ; 9: 2185, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319638

RESUMEN

Infiltrating immune cells are a key component of the tumor microenvironment and play central roles in dictating tumor fate, either promoting anti-tumor immune responses, or sustaining tumor growth, angiogenesis and metastasis. A distinctive microenvironment is often associated to different tumor types, with substantial differences in prognosis. The production of a variety of chemotactic factors by cancer and stromal cells orchestrates cell recruitment, local immune responses or cancer progression. In the last decades, different studies have highlighted how chemotactic cues, and in particular chemokines, can act as natural antagonists or induce synergistic effects on selective receptors by forming heterocomplexes, thus shaping migratory responses of immune cells. A variety of chemokines has been described to be able to form heterocomplexes both in vitro and in vivo under inflammatory conditions, but nowadays little is known on the presence and relevance of heterocomplexes in the tumor microenvironment. In recent years, the alarmin HMGB1, which can be massively released within the tumor microenvironment, has also been described to form a complex with the chemokine CXCL12 enhancing CXCR4-mediated signaling, thus providing an additional regulation of the activity of the chemokine system. In the present review, we will discuss the current knowledge on the synergy occurring between chemokines or inflammatory molecules, and describe the multiple functions exerted by the chemokines expressed in the tumor microenvironment, pointing our attention to the synergism as a possible modulator of tumor suppression or progression.


Asunto(s)
Quimiocina CXCL12/inmunología , Proteína HMGB1/inmunología , Neoplasias/inmunología , Multimerización de Proteína/inmunología , Microambiente Tumoral/inmunología , Quimiocina CXCL12/metabolismo , Quimiotaxis/inmunología , Progresión de la Enfermedad , Proteína HMGB1/metabolismo , Humanos , Leucocitos/inmunología , Leucocitos/metabolismo , Neoplasias/patología , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Transducción de Señal/inmunología
14.
Front Immunol ; 9: 2118, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30283452

RESUMEN

Chemokine synergy-inducing molecules are emerging as regulating factors in cell migration. The alarmin HMGB1, in its reduced form, can complex with CXCL12 enhancing its activity on monocytes via the chemokine receptor CXCR4, while the form containing a disulfide bond, by binding to TLR2 or TLR4, initiates a cascade of events leading to production of cytokines and chemokines. So far, the possibility that the CXCL12/HMGB1 heterocomplex could be maintained in chronic inflammation was debated, due to the release of reactive oxygen species. Therefore, we have assessed if the heterocomplex could remain active in Rheumatoid Arthritis (RA) and its relevance in the disease assessment. Monocytes from RA patients with active disease require a low concentration of HMGB1 to enhance CXCL12-induced migration, in comparison to monocytes from patients in clinical remission or healthy donors. The activity of the heterocomplex depends on disease activity, on the COX2 and JAK/STAT pathways, and is determined by the redox potential of the microenvironment. In RA, the presence of an active thioredoxin system correlates with the enhanced cell migration, and with the presence of the heterocomplex in the synovial fluid. The present study highlights how, in an unbalanced microenvironment, the activity of the thioredoxin system plays a crucial role in sustaining inflammation. Prostaglandin E2 stimulation of monocytes from healthy donors is sufficient to recapitulate the response observed in patients with active RA. The activation of mechanisms counteracting the oxidative stress in the extracellular compartment preserves HMGB1 in its reduced form, and contributes to fuel the influx of inflammatory cells. Targeting the heterocomplex formation and its activity could thus be an additional tool for dampening the inflammation sustained by cell recruitment, for those patients with chronic inflammatory conditions who poorly respond to current therapies.


Asunto(s)
Artritis Reumatoide/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Proteína HMGB1/farmacología , Monocitos/efectos de los fármacos , Adulto , Anciano , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Movimiento Celular/inmunología , Células Cultivadas , Dinoprostona/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Oxidación-Reducción , Unión Proteica/efectos de los fármacos , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
15.
J Leukoc Biol ; 99(6): 851-5, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26715684

RESUMEN

Directed migration and arrest of leukocytes during homeostasis, inflammation, and tumor development is mediated by the chemokine system, which governs leukocyte migration and activities. Although we understand well the effects of different chemokines one by one, much less was known about the potential consequences of the concomitant expression of multiple chemokines or of their interaction with inflammatory molecules on leukocyte migration and functions. In the past 10 yr, several studies revealed the existence of additional features of chemokines: they can antagonize chemokine receptors or synergize with other chemokines, also by forming heterocomplexes. Moreover, recent data show that not only chemokines but also the alarmin high-mobility group box 1 can for a complex with CXCL12, enhancing its potency on CXCR4. The molecular mechanism underlying the effect of the heterocomplex has been partially elucidated, whereas its structure is a matter of current investigations. The present review discusses the current knowledge and relevance of the functions of heterocomplexes formed between chemokines or between the chemokine CXCL12 and the alarmin high-mobility group box 1. These studies highlight the importance of taking into account, when approaching innovative therapies targeting the chemokine system, also the fact that some chemokines and molecules released in inflammation, can considerably affect the activity of chemokine receptor agonists.


Asunto(s)
Movimiento Celular , Quimiocinas/metabolismo , Animales , Proteína HMGB1/metabolismo , Humanos , Modelos Biológicos , Complejos Multiproteicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA