Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
RMD Open ; 10(1)2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38395454

RESUMEN

OBJECTIVES: Ankylosing spondylitis (AS) is a chronic inflammatory rheumatic disease affecting mainly the axial skeleton. Peripheral involvement (arthritis, enthesitis and dactylitis) and extra-musculoskeletal manifestations, including uveitis, psoriasis and bowel inflammation, occur in a relevant proportion of patients. AS is responsible for chronic and severe back pain caused by local inflammation that can lead to osteoproliferation and ultimately spinal fusion. The association of AS with the human leucocyte antigen-B27 gene, together with elevated levels of chemokines, CCL17 and CCL22, in the sera of patients with AS, led us to study the role of CCR4+ T cells in the disease pathogenesis. METHODS: CD8+CCR4+ T cells isolated from the blood of patients with AS (n=76) or healthy donors were analysed by multiparameter flow cytometry, and gene expression was evaluated by RNA sequencing. Patients with AS were stratified according to the therapeutic regimen and current disease score. RESULTS: CD8+CCR4+ T cells display a distinct effector phenotype and upregulate the inflammatory chemokine receptors CCR1, CCR5, CX3CR1 and L-selectin CD62L, indicating an altered migration ability. CD8+CCR4+ T cells expressing CX3CR1 present an enhanced cytotoxic profile, expressing both perforin and granzyme B. RNA-sequencing pathway analysis revealed that CD8+CCR4+ T cells from patients with active disease significantly upregulate genes promoting osteogenesis, a core process in AS pathogenesis. CONCLUSIONS: Our results shed light on a new molecular mechanism by which T cells may selectively migrate to inflammatory loci, promote new bone formation and contribute to the pathological ossification process observed in AS.


Asunto(s)
Espondilitis Anquilosante , Humanos , Espondilitis Anquilosante/genética , Espondilitis Anquilosante/metabolismo , Osteogénesis/genética , Subgrupos de Linfocitos T/metabolismo , Linfocitos T CD8-positivos/metabolismo , Inflamación
2.
Front Immunol ; 14: 1176619, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37251376

RESUMEN

Leukocyte trafficking is mainly governed by chemokines, chemotactic cytokines, which can be concomitantly produced in tissues during homeostatic conditions or inflammation. After the discovery and characterization of the individual chemokines, we and others have shown that they present additional properties. The first discoveries demonstrated that some chemokines act as natural antagonists on chemokine receptors, and prevent infiltration of leukocyte subsets in tissues. Later on it was shown that they can exert a repulsive effect on selective cell types, or synergize with other chemokines and inflammatory mediators to enhance chemokine receptors activities. The relevance of the fine-tuning modulation has been demonstrated in vivo in a multitude of processes, spanning from chronic inflammation to tissue regeneration, while its role in the tumor microenvironment needs further investigation. Moreover, naturally occurring autoantibodies targeting chemokines were found in tumors and autoimmune diseases. More recently in SARS-CoV-2 infection, the presence of several autoantibodies neutralizing chemokine activities distinguished disease severity, and they were shown to be beneficial, protecting from long-term sequelae. Here, we review the additional properties of chemokines that influence cell recruitment and activities. We believe these features need to be taken into account when designing novel therapeutic strategies targeting immunological disorders.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Quimiocinas/metabolismo , Inflamación , Receptores de Quimiocina/metabolismo , Autoanticuerpos
3.
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
4.
bioRxiv ; 2022 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-35664993

RESUMEN

Infection by SARS-CoV-2 leads to diverse symptoms, which can persist for months. While antiviral antibodies are protective, those targeting interferons and other immune factors are associated with adverse COVID-19 outcomes. Instead, we discovered that antibodies against specific chemokines are omnipresent after COVID-19, associated with favorable disease, and predictive of lack of long COVID symptoms at one year post infection. Anti-chemokine antibodies are present also in HIV-1 infection and autoimmune disorders, but they target different chemokines than those in COVID-19. Monoclonal antibodies derived from COVID- 19 convalescents that bind to the chemokine N-loop impair cell migration. Given the role of chemokines in orchestrating immune cell trafficking, naturally arising anti-chemokine antibodies associated with favorable COVID-19 may be beneficial by modulating the inflammatory response and thus bear therapeutic potential. One-Sentence Summary: Naturally arising anti-chemokine antibodies associate with favorable COVID-19 and predict lack of long COVID.

5.
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
6.
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
7.
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.

8.
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
9.
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
10.
J Leukoc Biol ; 104(2): 295-300, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29668065

RESUMEN

The activity of chemokines is regulated by several mechanisms that control the final cellular response. The present review discusses the complexity of the regulation of the chemokine system, and the novel findings describing how in persistent infections, the expression of chemokine receptors on the surface of T cells does not correlate with their homing potential. Thanks to the latest advances in our comprehension of the chemokine system, novel approaches targeting chemokines, chemokine receptors, or protein of their signaling pathway should be considered in order to achieve a personalized therapy.


Asunto(s)
Quimiocinas , Animales , Humanos
11.
Front Immunol ; 8: 1563, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29204145

RESUMEN

Vaccinology aims to understand what factors drive vaccine-induced immunity and protection. For many vaccines, however, the mechanisms underlying immunity and protection remain incompletely characterized at best, and except for neutralizing antibodies induced by viral vaccines, few correlates of protection exist. Recent omics and systems biology big data platforms have yielded valuable insights in these areas, particularly for viral vaccines, but in the case of more complex vaccines against bacterial infectious diseases, understanding is fragmented and limited. To fill this gap, the EC supported ADITEC project (http://www.aditecproject.eu/; http://stm.sciencemag.org/content/4/128/128cm4.full) featured a work package on "Molecular signatures of immunity and immunogenicity," aimed to identify key molecular mechanisms of innate and adaptive immunity during effector and memory stages of immune responses following vaccination. Specifically, technologies were developed to assess the human immune response to vaccination and infection at the level of the transcriptomic and proteomic response, T-cell and B-cell memory formation, cellular trafficking, and key molecular pathways of innate immunity, with emphasis on underlying mechanisms of protective immunity. This work intersected with other efforts in the ADITEC project. This review summarizes the main achievements of the work package.

12.
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
13.
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
14.
Front Immunol ; 7: 95, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27014274

RESUMEN

Prior investigations show that signaling activation through pattern recognition receptors can directly impact a number of inflammatory lung diseases. While toll-like receptor (TLR) 7 agonists have raised interest for their ability to inhibit allergen-induced pathological changes in experimental asthma conditions, the putative benefit of this treatment is limited by adverse effects. Our aim was to evaluate the therapeutic potential of two PEGylated purine-like compounds, TMX-302 and TMX-306, characterized by TLR7 partial agonistic activity; therefore, the compounds are expected to induce lower local and systemic adverse reactions. In vitro approaches and translation to murine models of obstructive and restrictive lung diseases were explored. In vitro studies with human PBMCs showed that both TMX-302 and TMX-306 marginally affects cytokine production as compared with equivalent concentrations of the TLR7 full agonist, TMX-202. The PEGylated compounds did not induce monocyte-derived DC maturation or B cell proliferation, differently from what observed after stimulation with TMX-202. Impact of PEGylated ligands on lung function and inflammatory changes was studied in animal models of acute lung injury, asthma, and silicosis following Lipopolysaccharide (LPS), allergen (ovalbumin), and silica inhalation, respectively. Subcutaneous injection of TMX-302 prevented LPS- and allergen-induced airway hyper-reactivity (AHR), leukocyte infiltration, and production of pro-inflammatory cytokines in the lung. However, intranasal instillation of TMX-302 led to neutrophil infiltration and failed to prevent allergen-induced AHR, despite inhibiting leukocyte counts in the BAL. Aerosolized TMX-306 given prophylactically, but not therapeutically, inhibited pivotal asthma features. Interventional treatment with intranasal instillation of TMX-306 significantly reduced the pulmonary fibrogranulomatous response and the number of silica particles in lung interstitial space in silicotic mice. These findings highlight the potential of TMX-306, emphasizing its value in drug development for lung diseases, and particularly silicosis.

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
16.
Genes Chromosomes Cancer ; 54(8): 516-526, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26052821

RESUMEN

The pathways of NOTCH and PI3K/AKT are dysregulated in about 60% and 48% of T-cell acute lymphoblastic leukemia (T-ALL) patients, respectively. In this context, they interact and cooperate in controlling tumor cell biology. Here, we propose a novel mechanism by which the PI3K/AKT pathway regulates NOTCH1 in T-ALL, starting from the evidence that the inhibition of PI3K/AKT signaling induced by treatment with LY294002 or transient transfection with a dominant negative AKT mutant downregulates NOTCH1 protein levels and activity, without affecting NOTCH1 transcription. We showed that the withdrawal of PI3K/AKT signaling was associated to NOTCH1 phosphorylation in tyrosine residues and monoubiquitination of NOTCH1 detected by Ubiquitin capture assay. Co-immunoprecipitation assay and colocalization analysis further showed that the E3 ubiquitin ligase c-Cbl interacts and monoubiquitinates NOTCH1, activating its lysosomal degradation. These results suggest that the degradation of NOTCH1 could represent a mechanism of control by which NOTCH1 receptors are actively removed from the cell surface. This mechanism is finely regulated by the PI3K/AKT pathway in physiological conditions. In pathological conditions characterized by PI3K/AKT hyperactivation, such as T-ALL, the excessive AKT signaling could lead to NOTCH1 signaling dysregulation. Therefore, a therapeutic strategy directed to PI3K/AKT in T-ALL could contemporaneously inhibit the dysregulated NOTCH1 signaling. © 2015 Wiley Periodicals, Inc.

17.
Vaccine ; 30(1): 78-94, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22037204

RESUMEN

The role of antibodies directed against the hyper variable envelope region V1 of human immunodeficiency virus type 1 (HIV-1), has not been thoroughly studied. We show that a vaccine able to elicit strain-specific non-neutralizing antibodies to this region of gp120 is associated with control of highly pathogenic chimeric SHIV(89.6P) replication in rhesus macaques. The vaccinated animal that had the highest titers of antibodies to the amino terminus portion of V1, prior to challenge, had secondary antibody responses that mediated cell killing by antibody-dependent cellular cytotoxicity (ADCC), as early as 2 weeks after infection and inhibited viral replication by antibody-dependent cell-mediated virus inhibition (ADCVI), by 4 weeks after infection. There was a significant inverse correlation between virus level and binding antibody titers to the envelope protein, (R=-0.83, p=0.015), and ADCVI (R=-0.84 p=0.044). Genotyping of plasma virus demonstrated in vivo selection of three SHIV(89.6P) variants with changes in potential N-linked glycosylation sites in V1. We found a significant inverse correlation between virus levels and titers of antibodies that mediated ADCVI against all the identified V1 virus variants. A significant inverse correlation was also found between neutralizing antibody titers to SHIV(89.6) and virus levels (R=-0.72 p=0.0050). However, passive inoculation of purified immunoglobulin from animal M316, the macaque that best controlled virus, to a naïve macaque, resulted in a low serum neutralizing antibodies and low ADCVI activity that failed to protect from SHIV(89.6P) challenge. Collectively, while our data suggest that anti-envelope antibodies with neutralizing and non-neutralizing Fc(R-dependent activities may be important in the control of SHIV replication, they also demonstrate that low levels of these antibodies alone are not sufficient to protect from infection.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/inmunología , Proteína gp120 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Vacunas contra el SIDA/administración & dosificación , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Humanos , Macaca mulatta , Carga Viral
18.
Blood ; 118(6): 1549-59, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21677314

RESUMEN

Disease development in human T-cell leukemia virus type 1 (HTLV-1)-infected individuals is positively correlated with the level of integrated viral DNA in T cells. HTLV-1 replication is positively regulated by Tax and Rex and negatively regulated by the p30 and HBZ proteins. In the present study, we demonstrate that HTLV-1 encodes another negative regulator of virus expression, the p13 protein. Expressed separately, p13 localizes to the mitochondria, whereas in the presence of Tax, part of it is ubiquitinated, stabilized, and rerouted to the nuclear speckles. The p13 protein directly binds Tax, decreases Tax binding to the CBP/p300 transcriptional coactivator, and, by reducing Tax transcriptional activity, suppresses viral expression. Because Tax stabilizes its own repressor, these findings suggest that HTLV-1 has evolved a complex mechanism to control its own replication. Further, these results highlight the importance of studying the function of the HTLV-1 viral proteins, not only in isolation, but also in the context of full viral replication.


Asunto(s)
Núcleo Celular/metabolismo , Productos del Gen tax/metabolismo , Virus Linfotrópico T Tipo 1 Humano/fisiología , Proteínas de los Retroviridae/metabolismo , Replicación Viral/fisiología , Western Blotting , Línea Celular , Regulación Viral de la Expresión Génica , Productos del Gen tax/genética , Células HEK293 , Células HeLa , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Inmunoprecipitación , Microscopía Confocal , Membranas Mitocondriales/metabolismo , Unión Proteica , Proteínas de los Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Replicación Viral/genética , Factores de Transcripción p300-CBP/metabolismo
19.
J Infect Dis ; 203(8): 1043-53, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21450994

RESUMEN

The licensed smallpox vaccine, ACAM2000, is a cell culture derivative of Dryvax. Both ACAM2000 and Dryvax are administered by skin scarification and can cause progressive vaccinia, with skin lesions that disseminate to distal sites. We have investigated the immunologic basis of the containment of vaccinia in the skin with the goal to identify safer vaccines for smallpox. Macaques were depleted systemically of T or B cells and vaccinated with either Dryvax or an attenuated vaccinia vaccine, LC16m8. B cell depletion did not affect the size of skin lesions induced by either vaccine. However, while depletion of both CD4(+) and CD8(+) T cells had no adverse effects on LC16m8-vaccinated animals, it caused progressive vaccinia in macaques immunized with Dryvax. As both Dryvax and LC16m8 vaccines protect healthy macaques from a lethal monkeypox intravenous challenge, our data identify LC16m8 as a safer and effective alternative to ACAM2000 and Dryvax vaccines for immunocompromised individuals.


Asunto(s)
Linfocitos B/fisiología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Piel/patología , Vacuna contra Viruela/efectos adversos , Animales , Anticuerpos Neutralizantes/sangre , Proteínas de Unión al Calcio , Depleción Linfocítica , Macaca mulatta , Mpox/mortalidad , Mpox/prevención & control , Vacuna contra Viruela/inmunología
20.
Blood ; 116(19): 3809-17, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-20647569

RESUMEN

The identification of the genes necessary for human T-cell leukemia virus (HTLV-1) persistence in humans may provide targets for therapeutic approaches. We demonstrate that ablation of the HTLV-1 genes encoding p12, p30, or the HBZ protein, does not affect viral infectivity in rabbits and in this species, only the absence of HBZ is associated with a consistent reduction in virus levels. We observed reversion of the HTLV-1 mutants to the HTLV-1 wild-type genotype in none of the inoculated rabbits. In contrast, in macaques, the absence of HBZ was associated with reversion of the mutant virus to the wild-type genotype in 3 of the 4 animals within weeks from infection. Similarly, reversion to the wild type was observed in 2 of the 4 macaque inoculated with the p30 mutant. The 4 macaques exposed to the p12 knock remained seronegative, and only 2 animals were positive at a single time point for viral DNA in tissues. Interestingly, we found that the p12 and the p30 mutants were also severely impaired in their ability to replicate in human dendritic cells. These data suggest that infection of dendritic cells may be required for the establishment and maintenance of HTLV-1 infection in primate species.


Asunto(s)
Células Dendríticas/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/fisiología , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Línea Celular , Cartilla de ADN/genética , ADN Viral/genética , Células Dendríticas/inmunología , Femenino , Eliminación de Gen , Genes Virales , Genes pX , Genotipo , Infecciones por HTLV-I/inmunología , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/inmunología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Humanos , Técnicas In Vitro , Macaca mulatta , Mutagénesis , Mutación , Conejos , Especificidad de la Especie , Linfocitos T/inmunología , Linfocitos T/virología , Proteínas Virales/genética , Proteínas Virales/fisiología , Virulencia/genética , Virulencia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...