Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Mol Med ; 28(1): 108, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-36071400

RESUMEN

BACKGROUND: High-mobility group box 1 protein (HMGB1) is an ubiquitous nuclear protein that once released in the extracellular space acts as a Damage Associated Molecular Pattern and promotes inflammation. HMGB1 is significantly elevated during Pseudomonas aeruginosa infections and has a clinical relevance in respiratory diseases such as Cystic Fibrosis (CF). Salicylates are HMGB1 inhibitors. To address pharmacological inhibition of HMGB1 with small molecules, we explored the therapeutic potential of pamoic acid (PAM), a salicylate with limited ability to cross epithelial barriers. METHODS: PAM binding to HMGB1 and CXCL12 was tested by Nuclear Magnetic Resonance Spectroscopy using chemical shift perturbation methods, and inhibition of HMGB1·CXCL12-dependent chemotaxis was investigated by cell migration experiments. Aerosol delivery of PAM, with single or repeated administrations, was tested in murine models of acute and chronic P. aeruginosa pulmonary infection in C57Bl/6NCrlBR mice. PAM efficacy was evaluated by read-outs including weight loss, bacterial load and inflammatory response in lung and bronco-alveolar lavage fluid. RESULTS: Our data and three-dimensional models show that PAM is a direct ligand of both HMGB1 and CXCL12. We also showed that PAM is able to interfere with heterocomplex formation and the related chemotaxis in vitro. Importantly, PAM treatment by aerosol was effective in reducing acute and chronic airway murine inflammation and damage induced by P. aeruginosa. The results indicated that PAM reduces leukocyte recruitment in the airways, in particular neutrophils, suggesting an impaired in vivo chemotaxis. This was associated with decreased myeloperoxidase and neutrophil elastase levels. Modestly increased bacterial burdens were recorded with single administration of PAM in acute infection; however, repeated administration in chronic infection did not affect bacterial burdens, indicating that the interference of PAM with the immune system has a limited risk of pulmonary exacerbation. CONCLUSIONS: This work established the efficacy of treating inflammation in chronic respiratory diseases, including bacterial infections, by topical delivery in the lung of PAM, an inhibitor of HMGB1.


Asunto(s)
Quimiocina CXCL12 , Proteína HMGB1 , Naftoles , Neumonía Bacteriana , Animales , Quimiocina CXCL12/antagonistas & inhibidores , Quimiotaxis/efectos de los fármacos , Modelos Animales de Enfermedad , Proteína HMGB1/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Naftoles/farmacología , Neumonía Bacteriana/tratamiento farmacológico , Pseudomonas aeruginosa/metabolismo
2.
EMBO Rep ; 20(10): e47788, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31418171

RESUMEN

Extracellular HMGB1 triggers inflammation following infection or injury and supports tumorigenesis in inflammation-related malignancies. HMGB1 has several redox states: reduced HMGB1 recruits inflammatory cells to injured tissues forming a heterocomplex with CXCL12 and signaling via its receptor CXCR4; disulfide-containing HMGB1 binds to TLR4 and promotes inflammatory responses. Here we show that diflunisal, an aspirin-like nonsteroidal anti-inflammatory drug (NSAID) that has been in clinical use for decades, specifically inhibits in vitro and in vivo the chemotactic activity of HMGB1 at nanomolar concentrations, at least in part by binding directly to both HMGB1 and CXCL12 and disrupting their heterocomplex. Importantly, diflunisal does not inhibit TLR4-dependent responses. Our findings clarify the mode of action of diflunisal and open the way to the rational design of functionally specific anti-inflammatory drugs.


Asunto(s)
Quimiocina CXCL12/metabolismo , Diflunisal/farmacología , Proteína HMGB1/metabolismo , Leucocitos/metabolismo , Células 3T3 , Animales , Quimiotaxis/efectos de los fármacos , Diflunisal/química , Disulfuros/metabolismo , Ácido Glicirrínico/farmacología , Humanos , Inflamación/patología , Leucocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Espectroscopía de Resonancia Magnética , Ratones
3.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2693-2704, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28716707

RESUMEN

Myocardial infarction (MI) is a major health burden worldwide. Extracellular High mobility group box 1 (HMGB1) regulates tissue healing after injuries. The reduced form of HMGB1 (fr-HMGB1) exerts chemotactic activity by binding CXCL12 through CXCR4, while the disulfide form, (ds-HMGB1), induces cytokines expression by TLR4. Here, we assessed the role of HMGB1 redox forms and the non-oxidizable mutant (3S) on human cardiac fibroblast (hcFbs) functions and cardiac remodeling after infarction. Among HMGB1 receptors, hcFbs express CXCR4. Fr-HMGB1 and 3S, but not ds-HMGB1, promote hcFbs migration through Src activation, while none of HMGB1 redox forms induces proliferation or inflammatory mediators. 3S is more effective than fr-HMGB1 in stimulating hcFbs migration and Src phosphorylation being active at lower concentrations and in oxidizing conditions. Notably, chemotaxis toward both proteins is CXCR4-dependent but, in contrast to fr-HMGB1, 3S does not require CXCL12 since hcFbs migration persists in the presence of the CXCL12/CXCR4 inhibitor AMD3100 or an anti-CXCL12 antibody. Interestingly, 3S interacts with CXCR4 and induces a different receptor conformation than CXCL12. Mice undergoing MI and receiving 3S exhibit adverse LV remodeling owing to an excessive collagen deposition promoted by a higher number of myofibroblasts. On the contrary, fr-HMGB1 ameliorates cardiac performance enhancing neoangiogenesis and reducing the infarcted area and fibrosis. Altogether, our results demonstrate that non-oxidizable HMGB1 induce a sustained cardiac fibroblasts migration despite the redox state of the environment and by altering CXCL12/CXCR4 axis. This affects proper cardiac remodeling after an infarction.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/metabolismo , Fibroblastos/metabolismo , Proteína HMGB1/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptores CXCR4/metabolismo , Femenino , Fibroblastos/patología , Humanos , Masculino , Infarto del Miocardio/patología , Miocardio/patología , Oxidación-Reducción
4.
Mol Med ; 21: 526-35, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26101955

RESUMEN

Salicylic acid (SA) and its derivatives have been used for millennia to reduce pain, fever and inflammation. In addition, prophylactic use of acetylsalicylic acid, commonly known as aspirin, reduces the risk of heart attack, stroke and certain cancers. Because aspirin is rapidly de-acetylated by esterases in human plasma, much of aspirin's bioactivity can be attributed to its primary metabolite, SA. Here we demonstrate that human high mobility group box 1 (HMGB1) is a novel SA-binding protein. SA-binding sites on HMGB1 were identified in the HMG-box domains by nuclear magnetic resonance (NMR) spectroscopic studies and confirmed by mutational analysis. Extracellular HMGB1 is a damage-associated molecular pattern molecule (DAMP), with multiple redox states. SA suppresses both the chemoattractant activity of fully reduced HMGB1 and the increased expression of proinflammatory cytokine genes and cyclooxygenase 2 (COX-2) induced by disulfide HMGB1. Natural and synthetic SA derivatives with greater potency for inhibition of HMGB1 were identified, providing proof-of-concept that new molecules with high efficacy against sterile inflammation are attainable. An HMGB1 protein mutated in one of the SA-binding sites identified by NMR chemical shift perturbation studies retained chemoattractant activity, but lost binding of and inhibition by SA and its derivatives, thereby firmly establishing that SA binding to HMGB1 directly suppresses its proinflammatory activities. Identification of HMGB1 as a pharmacological target of SA/aspirin provides new insights into the mechanisms of action of one of the world's longest and most used natural and synthetic drugs. It may also provide an explanation for the protective effects of low-dose aspirin usage.


Asunto(s)
Aspirina/farmacología , Proteína HMGB1/genética , Inflamación/genética , Ácido Salicílico/farmacología , Aspirina/química , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Proteína HMGB1/biosíntesis , Proteína HMGB1/química , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Mutación , Resonancia Magnética Nuclear Biomolecular , Ácido Salicílico/química
5.
Nat Commun ; 15(1): 1201, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38331917

RESUMEN

Chemokine heterodimers activate or dampen their cognate receptors during inflammation. The CXCL12 chemokine forms with the fully reduced (fr) alarmin HMGB1 a physiologically relevant heterocomplex (frHMGB1•CXCL12) that synergically promotes the inflammatory response elicited by the G-protein coupled receptor CXCR4. The molecular details of complex formation were still elusive. Here we show by an integrated structural approach that frHMGB1•CXCL12 is a fuzzy heterocomplex. Unlike previous assumptions, frHMGB1 and CXCL12 form a dynamic equimolar assembly, with structured and unstructured frHMGB1 regions recognizing the CXCL12 dimerization surface. We uncover an unexpected role of the acidic intrinsically disordered region (IDR) of HMGB1 in heterocomplex formation and its binding to CXCR4 on the cell surface. Our work shows that the interaction of frHMGB1 with CXCL12 diverges from the classical rigid heterophilic chemokines dimerization. Simultaneous interference with multiple interactions within frHMGB1•CXCL12 might offer pharmacological strategies against inflammatory conditions.


Asunto(s)
Quimiocina CXCL12 , Proteína HMGB1 , Humanos , Quimiocina CXCL12/metabolismo , Proteína HMGB1/metabolismo , Receptores CXCR4/metabolismo , Inflamación , Transducción de Señal
6.
J Cell Biol ; 179(1): 33-40, 2007 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-17923528

RESUMEN

Tissue damage is usually followed by healing, as both differentiated and stem cells migrate to replace dead or damaged cells. Mesoangioblasts (vessel-associated stem cells that can repair muscles) and fibroblasts migrate toward soluble factors released by damaged tissue. Two such factors are high mobility group box 1 (HMGB1), a nuclear protein that is released by cells undergoing unscheduled death (necrosis) but not by apoptotic cells, and stromal derived factor (SDF)-1/CXCL12. We find that HMGB1 activates the canonical nuclear factor kappaB (NF-kappaB) pathway via extracellular signal-regulated kinase phosphorylation. NF-kappaB signaling is necessary for chemotaxis toward HMGB1 and SDF-1/CXCL12, but not toward growth factor platelet-derived growth factor, formyl-met-leu-phe (a peptide that mimics bacterial invasion), or the archetypal NF-kappaB-activating signal tumor necrosis factor alpha. In dystrophic mice, mesoangioblasts injected into the general circulation ingress inefficiently into muscles if their NF-kappaB signaling pathway is disabled. These findings suggest that NF-kappaB signaling controls tissue regeneration in addition to early events in inflammation.


Asunto(s)
Quimiotaxis/fisiología , Proteína HMGB1/metabolismo , FN-kappa B/fisiología , Transducción de Señal , Animales , Línea Celular , Quimiocina CXCL12/metabolismo , Fibroblastos/metabolismo , Fibroblastos/fisiología , Proteínas Fluorescentes Verdes/análisis , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Necrosis , Fosforilación , Proteínas Recombinantes de Fusión/análisis
7.
Mol Ther ; 19(2): 266-73, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20924364

RESUMEN

Previous data report that fibroblast growth factor-2 (FGF-2)-derived peptide FREG potently inhibits FGF-2-dependent angiogenesis in vitro and in vivo. Here, we show that FREG inhibits up to 70% in vitro growth and invasion/migration of smooth muscle and melanoma cells. Such inhibition is mediated by platelet-derived growth factor-receptor-α (PDGF-Rα); in fact, proliferation and migration were restored upon PDGF-Rα neutralization. Further experiments demonstrated that FREG interacts with PDGF-Rα both in vitro and in vivo and stimulates its phosphorylation. We have previously shown that overexpressing PDGF-Rα strongly inhibits melanoma growth in vivo; we, therefore, hypothesized that PDGF-Rα agonists may represent a novel tool to inhibit melanoma growth in vivo. To support this hypothesis, FREG was inoculated intravenously (i.v.) in a mouse melanoma model and markedly inhibited pulmonary metastases formation. Immunohistochemical analyses showed less proliferation, less angiogenesis, and more apoptosis in metastasized lungs upon FREG treatment, as compared to untreated controls. Finally, in preliminary acute toxicity studies, FREG showed no toxicity signs in healthy animals, and neither microscopic nor macroscopic toxicity at the liver, kidney, and lungs level. Altogether, these data indicate that FREG systemic treatment strongly inhibits melanoma metastases development and indicate for the first time that agonists of PDGF-Rα may control melanoma both in vitro and in vivo.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/química , Melanoma/tratamiento farmacológico , Péptidos/química , Péptidos/uso terapéutico , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa
8.
EMBO Mol Med ; 13(6): e12344, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33956406

RESUMEN

Boosting antitumor immunity has emerged as a powerful strategy in cancer treatment. While releasing T-cell brakes has received most attention, tumor recognition by T cells is a pre-requisite. Radiotherapy and certain cytotoxic drugs induce the release of damage-associated molecular patterns, which promote tumor antigen cross-presentation and T-cell priming. Antibodies against the "do not eat me" signal CD47 cause macrophage phagocytosis of live tumor cells and drive the emergence of antitumor T cells. Here we show that CXCR4 activation, so far associated only with tumor progression and metastasis, also flags tumor cells to immune recognition. Both CXCL12, the natural CXCR4 ligand, and BoxA, a fragment of HMGB1, promote the release of DAMPs and the internalization of CD47, leading to protective antitumor immunity. We designate as Immunogenic Surrender the process by which CXCR4 turns in tumor cells to macrophages, thereby subjecting a rapidly growing tissue to immunological scrutiny. Importantly, while CXCL12 promotes tumor cell proliferation, BoxA reduces it, and might be exploited for the treatment of malignant mesothelioma and a variety of other tumors.


Asunto(s)
Antígeno CD47 , Mesotelioma , Animales , Línea Celular Tumoral , Inmunización , Macrófagos , Mesotelioma/inmunología , Mesotelioma/metabolismo , Mesotelioma/terapia , Ratones , Fagocitosis
9.
Front Chem ; 8: 598710, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324614

RESUMEN

HMGB1 is a key molecule that both triggers and sustains inflammation following infection or injury, and is involved in a large number of pathologies, including cancer. HMGB1 participates in the recruitment of inflammatory cells, forming a heterocomplex with the chemokine CXCL12 (HMGB1·CXCL12), thereby activating the G-protein coupled receptor CXCR4. Thus, identification of molecules that disrupt this heterocomplex can offer novel pharmacological opportunities to treat inflammation-related diseases. To identify new HMGB1·CXCL12 inhibitors we have performed a study on the ligandability of the single HMG boxes of HMGB1 followed by a virtual screening campaign on both HMG boxes using Zbc Drugs and three different docking programs (Glide, AutoDock Vina, and AutoDock 4.2.6). The best poses in terms of scoring functions, visual inspection, and predicted ADME properties were further filtered according to a pharmacophore model based on known HMGB1 binders and clustered according to their structures. Eight compounds representative of the clusters were tested for HMGB1 binding by NMR. We identified 5,5'-methylenedi-2,3-cresotic acid (2a) as a binder of both HMGB1 and CXCL12; 2a also targets the HMGB1·CXCL12 heterocomplex. In cell migration assays 2a inhibited the chemotactic activity of HMGB1·CXCL12 with IC50 in the subnanomolar range, the best documented up to now. These results pave the way for future structure activity relationship studies to optimize the pharmacological targeting of HMGB1·CXCL12 for anti-inflammatory purposes.

10.
J Cell Biol ; 164(3): 441-9, 2004 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-14744997

RESUMEN

High mobility group box 1 (HMGB1) is an abundant chromatin protein that acts as a cytokine when released in the extracellular milieu by necrotic and inflammatory cells. Here, we show that extracellular HMGB1 and its receptor for advanced glycation end products (RAGE) induce both migration and proliferation of vessel-associated stem cells (mesoangioblasts), and thus may play a role in muscle tissue regeneration. In vitro, HMGB1 induces migration and proliferation of both adult and embryonic mesoangioblasts, and disrupts the barrier function of endothelial monolayers. In living mice, mesoangioblasts injected into the femoral artery migrate close to HMGB1-loaded heparin-Sepharose beads implanted in healthy muscle, but are unresponsive to control beads. Interestingly, alpha-sarcoglycan null dystrophic muscle contains elevated levels of HMGB1; however, mesoangioblasts migrate into dystrophic muscle even if their RAGE receptor is disabled. This implies that the HMGB1-RAGE interaction is sufficient, but not necessary, for mesoangioblast homing; a different pathway might coexist. Although the role of endogenous HMGB1 in the reconstruction of dystrophic muscle remains to be clarified, injected HMGB1 may be used to promote tissue regeneration.


Asunto(s)
División Celular/fisiología , Movimiento Celular/fisiología , Endotelio Vascular/metabolismo , Proteína HMGB1/metabolismo , Células Madre/fisiología , Animales , Bovinos , Trasplante de Células , Células Cultivadas , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Embrión de Mamíferos/fisiología , Endotelio Vascular/citología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/metabolismo , Regeneración/fisiología , Sarcoglicanos , Células Madre/citología
11.
Chem Biol ; 14(4): 431-41, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17462578

RESUMEN

High-mobility group box 1 protein (HMGB1) is a nuclear component, but extracellularly it serves as a signaling molecule involved in acute and chronic inflammation, for example in sepsis and arthritis. The identification of HMGB1 inhibitors is therefore of significant experimental and clinical interest. We show that glycyrrhizin, a natural anti-inflammatory and antiviral triterpene in clinical use, inhibits HMGB1 chemoattractant and mitogenic activities, and has a weak inhibitory effect on its intranuclear DNA-binding function. NMR and fluorescence studies indicate that glycyrrhizin binds directly to HMGB1 (K(d) approximately 150 microM), interacting with two shallow concave surfaces formed by the two arms of both HMG boxes. Our results explain in part the anti-inflammatory properties of glycyrrhizin, and might direct the design of new derivatives with improved HMGB1-binding properties.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Citocinas/antagonistas & inhibidores , Ácido Glicirrínico/metabolismo , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/metabolismo , Células 3T3 , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Sitios de Unión , Citocinas/metabolismo , ADN/metabolismo , Fluorescencia , Ácido Glicirrínico/química , Ácido Glicirrínico/farmacología , Proteína HMGB1/química , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Mitógenos/antagonistas & inhibidores , Mitógenos/farmacología , Modelos Moleculares , Unión Proteica
12.
J Exp Med ; 215(1): 303-318, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29203538

RESUMEN

Inflammation and tissue regeneration follow tissue damage, but little is known about how these processes are coordinated. High Mobility Group Box 1 (HMGB1) is a nuclear protein that, when released on injury, triggers inflammation. We previously showed that HMGB1 with reduced cysteines is a chemoattractant, whereas a disulfide bond makes it a proinflammatory cytokine. Here we report that fully reduced HMGB1 orchestrates muscle and liver regeneration via CXCR4, whereas disulfide HMGB1 and its receptors TLR4/MD-2 and RAGE (receptor for advanced glycation end products) are not involved. Injection of HMGB1 accelerates tissue repair by acting on resident muscle stem cells, hepatocytes, and infiltrating cells. The nonoxidizable HMGB1 mutant 3S, in which serines replace cysteines, promotes muscle and liver regeneration more efficiently than the wild-type protein and without exacerbating inflammation by selectively interacting with CXCR4. Overall, our results show that the reduced form of HMGB1 coordinates tissue regeneration and suggest that 3S may be used to safely accelerate healing after injury in diverse clinical contexts.


Asunto(s)
Proteína HMGB1/metabolismo , Regeneración Hepática/fisiología , Músculos/metabolismo , Músculos/fisiología , Receptores CXCR4/metabolismo , Animales , Línea Celular , Factores Quimiotácticos/metabolismo , Citocinas/metabolismo , Células HEK293 , Hepatocitos/metabolismo , Hepatocitos/fisiología , Humanos , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Cicatrización de Heridas/fisiología
13.
J Exp Med ; 209(3): 551-63, 2012 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-22370717

RESUMEN

After tissue damage, inflammatory cells infiltrate the tissue and release proinflammatory cytokines. HMGB1 (high mobility group box 1), a nuclear protein released by necrotic and severely stressed cells, promotes cytokine release via its interaction with the TLR4 (Toll-like receptor 4) receptor and cell migration via an unknown mechanism. We show that HMGB1-induced recruitment of inflammatory cells depends on CXCL12. HMGB1 and CXCL12 form a heterocomplex, which we characterized by nuclear magnetic resonance and surface plasmon resonance, that acts exclusively through CXCR4 and not through other HMGB1 receptors. Fluorescence resonance energy transfer data show that the HMGB1-CXCL12 heterocomplex promotes different conformational rearrangements of CXCR4 from that of CXCL12 alone. Mononuclear cell recruitment in vivo into air pouches and injured muscles depends on the heterocomplex and is inhibited by AMD3100 and glycyrrhizin. Thus, inflammatory cell recruitment and activation both depend on HMGB1 via different mechanisms.


Asunto(s)
Quimiocina CXCL12/fisiología , Proteína HMGB1/fisiología , Inflamación/etiología , Receptores CXCR4/fisiología , Animales , Secuencia de Bases , Señalización del Calcio , Movimiento Celular/fisiología , Quimiocina CXCL12/química , ADN Complementario/genética , Fibroblastos/fisiología , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Proteína HMGB1/química , Humanos , Inflamación/patología , Inflamación/fisiopatología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Monocitos/fisiología , Complejos Multiproteicos/química , Células 3T3 NIH , Resonancia Magnética Nuclear Biomolecular , Receptor para Productos Finales de Glicación Avanzada , Receptores CXCR4/química , Receptores CXCR4/genética , Receptores Inmunológicos/fisiología , Transducción de Señal , Resonancia por Plasmón de Superficie , Receptores Toll-Like/fisiología , Transfección
14.
J Exp Med ; 209(9): 1519-28, 2012 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-22869893

RESUMEN

Tissue damage causes inflammation, by recruiting leukocytes and activating them to release proinflammatory mediators. We show that high-mobility group box 1 protein (HMGB1) orchestrates both processes by switching among mutually exclusive redox states. Reduced cysteines make HMGB1 a chemoattractant, whereas a disulfide bond makes it a proinflammatory cytokine and further cysteine oxidation to sulfonates by reactive oxygen species abrogates both activities. We show that leukocyte recruitment and activation can be separated. A nonoxidizable HMGB1 mutant in which serines replace all cysteines (3S-HMGB1) does not promote cytokine production, but is more effective than wild-type HMGB1 in recruiting leukocytes in vivo. BoxA, a HMGB1 inhibitor, interferes with leukocyte recruitment but not with activation. We detected the different redox forms of HMGB1 ex vivo within injured muscle. HMGB1 is completely reduced at first and disulfide-bonded later. Thus, HMGB1 orchestrates both key events in sterile inflammation, leukocyte recruitment and their induction to secrete inflammatory cytokines, by adopting mutually exclusive redox states.


Asunto(s)
Citocinas/metabolismo , Proteína HMGB1/metabolismo , Inflamación/metabolismo , Leucocitos/citología , Animales , Anticuerpos Monoclonales/farmacología , Movimiento Celular/efectos de los fármacos , Factores Quimiotácticos/metabolismo , Cisteína/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/inmunología , Leucocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/lesiones , Mutación , Ratas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
15.
Autoimmunity ; 42(4): 308-10, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19811285

RESUMEN

High mobility group B box (HMGB) proteins are a family of chromatin proteins made up of two basic DNA binding domains, HMG box A and B, and a C-terminal acidic tail. HMGB have a highly conserved sequence, but different expression pattern: HMGB1 is almost ubiquitous, whereas the others are highly expressed in only a few tissues in adults. We previously demonstrated that HMGB1 is released by necrotic cells and has chemoattractant activity for inflammatory and stem cells, via binding to receptor for advanced glycation endproducts (RAGE). HMGB1 can be actively secreted by inflammatory cells. Here, we report that also HMGB2 can be secreted by THP-1 cells, and promotes proliferation and migration of endothelial cells. These functions of HMGB2 are exerted via engagement of RAGE, whose blockade completely abrogates cell responses. Since extracellular HMGB2 has been detected in the blood and other biological fluids, it might be necessary to target HMGB2 at the same time as HMGB1 for therapeutical efficacy.


Asunto(s)
Factores Quimiotácticos/metabolismo , Proteína HMGB1/metabolismo , Proteína HMGB2/metabolismo , Células Mieloides/metabolismo , Animales , Western Blotting , Movimiento Celular/inmunología , Proliferación Celular , Factores Quimiotácticos/inmunología , Proteína HMGB1/inmunología , Proteína HMGB2/inmunología , Humanos , Ratones , Ratones Noqueados , Células Mieloides/inmunología
16.
J Leukoc Biol ; 86(3): 617-23, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19401391

RESUMEN

HMGB1 is a nuclear protein that signals tissue damage, as it is released by cells dying traumatically or secreted by activated innate immunity cells. Extracellular HMGB1 elicits the migration to the site of tissue damage of several cell types, including inflammatory cells and stem cells. The identity of the signaling pathways activated by extracellular HMGB1 is not known completely: We reported previously that ERK and NF-kappaB pathways are involved, and we report here that Src is also activated. The ablation of Src or inhibition with the kinase inhibitor PP2 blocks migration toward HMGB1. Src associates to and mediates the phosphorylation of FAK and the formation of focal adhesions.


Asunto(s)
Movimiento Celular/fisiología , Fibroblastos/fisiología , Proteína HMGB1/metabolismo , Leucocitos Mononucleares/fisiología , Familia-src Quinasas/fisiología , Células 3T3 , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Quimiotaxis/fisiología , Materiales Biocompatibles Revestidos/metabolismo , Medio de Cultivo Libre de Suero , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibronectinas/metabolismo , Adhesiones Focales/metabolismo , Proteína HMGB1/farmacología , Humanos , Punto Isoeléctrico , Leucocitos Mononucleares/efectos de los fármacos , Ratones , Paxillin/metabolismo , Fosforilación/efectos de los fármacos , Pirimidinas/farmacología , Temperatura , Factores de Tiempo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
17.
J Exp Med ; 205(13): 3007-18, 2008 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-19064698

RESUMEN

Autoantibodies against double-stranded DNA (dsDNA) and nucleosomes represent a hallmark of systemic lupus erythematosus (SLE). However, the mechanisms involved in breaking the immunological tolerance against these poorly immunogenic nuclear components are not fully understood. Impaired phagocytosis of apoptotic cells with consecutive release of nuclear antigens may contribute to the immune pathogenesis. The architectural chromosomal protein and proinflammatory mediator high mobility group box protein 1 (HMGB1) is tightly attached to the chromatin of apoptotic cells. We demonstrate that HMGB1 remains bound to nucleosomes released from late apoptotic cells in vitro. HMGB1-nucleosome complexes were also detected in plasma from SLE patients. HMGB1-containing nucleosomes from apoptotic cells induced secretion of interleukin (IL) 1beta, IL-6, IL-10, and tumor necrosis factor (TNF) alpha and expression of costimulatory molecules in macrophages and dendritic cells (DC), respectively. Neither HMGB1-free nucleosomes from viable cells nor nucleosomes from apoptotic cells lacking HMGB1 induced cytokine production or DC activation. HMGB1-containing nucleosomes from apoptotic cells induced anti-dsDNA and antihistone IgG responses in a Toll-like receptor (TLR) 2-dependent manner, whereas nucleosomes from living cells did not. In conclusion, HMGB1-nucleosome complexes activate antigen presenting cells and, thereby, may crucially contribute to the pathogenesis of SLE via breaking the immunological tolerance against nucleosomes/dsDNA.


Asunto(s)
Proteína HMGB1/inmunología , Fenómenos del Sistema Inmunológico/fisiología , Inflamación/inmunología , Lupus Eritematoso Sistémico/inmunología , Nucleosomas/inmunología , Animales , Apoptosis/fisiología , Autoanticuerpos/inmunología , Línea Celular , Citocinas/inmunología , Femenino , Proteína HMGB1/genética , Humanos , Lupus Eritematoso Sistémico/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 9/inmunología
18.
Blood ; 99(6): 2045-53, 2002 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11877278

RESUMEN

Basic fibroblast growth factor (bFGF) and platelet-derived growth factor-BB (PDGF-BB) modulate vascular wall cell function in vitro and angiogenesis in vivo. The aim of the current study was to determine how bovine aorta endothelial cells (BAECs) respond to the simultaneous exposure to PDGF-BB and bFGF. It was found that bFGF-dependent BAEC migration, proliferation, and differentiation into tubelike structures on reconstituted extracellular matrix (Matrigel) were inhibited by PDGF-BB. The role played by PDGF receptor alpha (PDGF-Ralpha) was investigated by selective stimulation with PDGF-AA, by blocking PDGF-BB-binding to PDGF-Ralpha with neomycin, or by transfecting cells with dominant-negative forms of the receptors to selectively impair either PDGF-Ralpha or PDGF-Rbeta function. In all cases, PDGF-Ralpha impairment abolished the inhibitory effect of PDGF-BB on bFGF-directed BAEC migration. In addition, PDGF-Ralpha phosphorylation was increased in the presence of bFGF and PDGF, as compared to PDGF alone, whereas mitogen-activated protein kinase phosphorylation was decreased in the presence of PDGF-BB and bFGF compared with bFGF alone. In vivo experiments showed that PDGF-BB and PDGF-AA inhibited bFGF-induced angiogenesis in vivo in the chick embryo chorioallantoic membrane assay and that PDGF-BB inhibited bFGF-induced angiogenesis in Matrigel plugs injected subcutaneously in CD1 mice. Taken together these results show that PDGF inhibits the angiogenic properties of bFGF in vitro and in vivo, likely through PDGF-Ralpha stimulation.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Aorta/citología , Becaplermina , Bovinos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Embrión de Pollo , Antagonismo de Drogas , Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología
19.
Blood ; 103(11): 4180-7, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-14982875

RESUMEN

Peptides containing the Arg-Gly-Asp (RGD) motif inhibit cell adhesion and exhibit a variety of other biologic effects including anticoagulant and antimetastatic activities. The aim of the present study was to examine the anchorage-independent effects of an RGD-containing peptide, Arg-Gly-Asp-Ser (RGDS), on human umbilical vein endothelial cells (HUVECs). Assays were performed on HUVECs seeded onto collagen IV; under these experimental conditions RGDS did not exert antiadhesive effects but significantly reduced FGF-2-dependent chemotaxis after 4 hours of treatment and reduced proliferation after 24 hours of treatment. Experiments carried out with caspase-specific inhibitors indicated that the observed antichemotactic effects required caspase 8 and caspase 9 activation. RGDS activated both caspase 8 and caspase 9 after 4 hours of treatment and caspase 3 after 24 hours of treatment, and markedly enhanced HUVEC apoptosis by transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL)/Hoechst staining and fluorescence-activated cell sorting (FACS) analysis. Finally, confocal microscopy showed that RGDS localizes in the cytoplasm of live HUVECs within 4 hours and in vitro experiments showed that RGDS directly interacts with recombinant caspases 8 and 9 in a specific way. In summary, these results indicate that RGDS directly binds and activates caspases 8 and 9, inhibits chemotaxis, and induces apoptosis of HUVECs with a mechanism independent from its antiadhesive effect.


Asunto(s)
Caspasas/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Oligopéptidos/farmacología , Caspasa 8 , Caspasa 9 , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Activación Enzimática/efectos de los fármacos , Humanos , Oligopéptidos/metabolismo , Venas Umbilicales/citología
20.
J Biol Chem ; 278(10): 8751-60, 2003 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-12496262

RESUMEN

Fibroblast growth factor 2 (FGF-2) is a potent factor modulating the activity of many cell types. Its dimerization and binding to high affinity receptors are considered to be necessary steps to induce FGF receptor phosphorylation and signaling activation. A structural analysis was carried out and a region encompassing residues 48-58 of human FGF-2 was identified, as potentially involved in FGF-2 dimerization. A peptide (FREG-48-58) derived from this region strongly and specifically inhibited FGF-2 induced proliferation and migration of primary bovine aorta endothelial cells (BAEC) in vitro, and markedly reduced FGF-2-dependent angiogenesis in two distinct in vivo assays. To further investigate the role of region 48-58, a polyclonal antibody raised against FREG-(48-58) was tested and was found to block FGF-2 action in vitro. Human FGF-2 has three histidine residues, one falling within the region 48-58. Chemical modification of histidine residues blocked FGF-2 activity and FREG-(48-58) inhibitory effect in vitro, indicating that histidine residues, in particular the one within FREG-(48-58) region, play a crucial role in the observed activity. Additional experiments showed that FREG-(48-58) specifically interacted with FGF-2, impaired FGF-2-interaction with itself, with heparin and with FGF receptor 1, and inhibited FGF-2-induced receptor phosphorylation and FGF-2 internalization. These data indicate for the first time that region 48-58 of FGF-2 is a functional domain controlling FGF-2 activity.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Neovascularización Fisiológica/fisiología , Secuencia de Aminoácidos , Animales , Bovinos , Células Cultivadas , Embrión de Pollo , Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/química , Factor 2 de Crecimiento de Fibroblastos/genética , Histidina/metabolismo , Humanos , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA