Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Pharm ; 18(8): 3171-3180, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34279974

RESUMEN

Current treatment of chronic wounds has been critically limited by various factors, including bacterial infection, biofilm formation, impaired angiogenesis, and prolonged inflammation. Addressing these challenges, we developed a multifunctional wound dressing-based three-pronged approach for accelerating wound healing. The multifunctional wound dressing, composed of nanofibers, functional nanoparticles, natural biopolymers, and selected protein and peptide, can target multiple endogenous repair mechanisms and represents a promising alternative to current wound healing products.


Asunto(s)
Anexina A1/administración & dosificación , Antiinflamatorios/administración & dosificación , Vendajes , Diabetes Mellitus Experimental/complicaciones , Proteínas Relacionadas con la Folistatina/administración & dosificación , Péptidos/administración & dosificación , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Herida Quirúrgica/complicaciones , Herida Quirúrgica/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Infección de Heridas/complicaciones , Infección de Heridas/tratamiento farmacológico , Células 3T3 , Animales , Materiales Biocompatibles/administración & dosificación , Biopolímeros/química , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Células HaCaT , Humanos , Nanopartículas Magnéticas de Óxido de Hierro/química , Masculino , Ensayo de Materiales/métodos , Ratones , Nanofibras/química , Ratas , Ratas Wistar , Infecciones Estafilocócicas/microbiología , Resultado del Tratamiento , Infección de Heridas/microbiología
2.
Fish Shellfish Immunol ; 94: 27-37, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31465876

RESUMEN

In teleost fish, myelopoiesis is maintained both in the head (HK) and trunk kidney (TK), but only the HK holds the endocrine cells that produce the stress hormone cortisol. We now compared the effects of prolonged restraint stress (in vivo) and cortisol (in vitro) on the polarization of HK and TK-derived carp macrophages. Monocytes/macrophages from both sources were treated in vitro with cortisol, lipopolysaccharide or with both factors combined. In vivo, fish were challenged by a prolonged restraint stress. Gene expression of several markers typical for classical M1 and alternative M2 macrophage polarization, as well as glucocorticoid receptors, were measured. Cells from both sources did not differ in the constitutive gene expression of glucocorticoid receptors, whereas they significantly differed in their response to cortisol and stress. In the LPS-stimulated HK monocytes/macrophages, cortisol in vitro counteracted the action of LPS while the effects of cortisol on the activity of TK monocytes/macrophages were less explicit. In vivo, restraint stress up-regulated gene expression of M2 markers in freshly isolated HK monocytes/macrophages, while at the same time it did not affect TK monocytes/macrophages. Moreover, LPS-stimulated HK monocytes/macrophages from stressed animals showed only minor differences in the gene expression of M1 and M2 markers, compared to LPS-treated monocytes/macrophages from control fish. In contrast, stress-induced changes in TK-derived LPS-treated cells were more pronounced. However, these changes did not clearly indicate whether in TK monocytes/macrophages stress will stimulate classical or alternative polarization. Altogether, our results imply that cortisol in vitro and stress in vivo direct HK, but not TK, monocytes/macrophages to the path of alternative polarization. These findings reveal that like in mammals, also in fish the glucocorticoids form important stimulators of alternative macrophage polarization.


Asunto(s)
Anexina A1/administración & dosificación , Carpas/fisiología , Proteínas de Peces/administración & dosificación , Expresión Génica/inmunología , Hidrocortisona/administración & dosificación , Macrófagos/inmunología , Péptidos/administración & dosificación , Estrés Fisiológico/inmunología , Animales , Carpas/inmunología , Inflamación/inmunología , Inflamación/veterinaria , Macrófagos/metabolismo
3.
Brain Behav Immun ; 51: 212-222, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26321046

RESUMEN

Sex differences have been widely reported in neuroinflammatory disorders, focusing on the contributory role of estrogen. The microvascular endothelium of the brain is a critical component of the blood-brain barrier (BBB) and it is recognized as a major interface for communication between the periphery and the brain. As such, the cerebral capillary endothelium represents an important target for the peripheral estrogen neuroprotective functions, leading us to hypothesize that estrogen can limit BBB breakdown following the onset of peripheral inflammation. Comparison of male and female murine responses to peripheral LPS challenge revealed a short-term inflammation-induced deficit in BBB integrity in males that was not apparent in young females, but was notable in older, reproductively senescent females. Importantly, ovariectomy and hence estrogen loss recapitulated an aged phenotype in young females, which was reversible upon estradiol replacement. Using a well-established model of human cerebrovascular endothelial cells we investigated the effects of estradiol upon key barrier features, namely paracellular permeability, transendothelial electrical resistance, tight junction integrity and lymphocyte transmigration under basal and inflammatory conditions, modeled by treatment with TNFα and IFNγ. In all cases estradiol prevented inflammation-induced defects in barrier function, action mediated in large part through up-regulation of the central coordinator of tight junction integrity, annexin A1. The key role of this protein was then further confirmed in studies of human or murine annexin A1 genetic ablation models. Together, our data provide novel mechanisms for the protective effects of estrogen, and enhance our understanding of the beneficial role it plays in neurovascular/neuroimmune disease.


Asunto(s)
Anexina A1/fisiología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Estradiol/fisiología , Inflamación/inmunología , Inflamación/fisiopatología , Linfocitos/fisiología , Animales , Anexina A1/administración & dosificación , Movimiento Celular/efectos de los fármacos , Citocinas/metabolismo , Estradiol/administración & dosificación , Femenino , Humanos , Inflamación/inducido químicamente , Lipopolisacáridos , Linfocitos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
4.
J Autoimmun ; 58: 1-11, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25619792

RESUMEN

Annexin-A1 (Anx-A1) is an endogenous anti-inflammatory molecule and while described as a repressor of innate immune responses, the role of Anx-A1 in adaptive immunity, and in particular in T helper (Th) cell responses, remains controversial. We have used a T-cell mediated mouse model of retinal autoimmune disease to unravel the role of Anx-A1 in the development of autoreactive Th cell responses and pathology. RBP1-20-immunized C57BL/6 Anx-A1(-/-) mice exhibit significantly enhanced retinal inflammation and pathology as a result of an uncontrolled proliferation and activation of Th17 cells. This is associated with a limited capacity to induce SOCS3, resulting in un-restricted phosphorylation of STAT3. RBP1-20-specific CD4(+) cells from immunized Anx-A1(-/-) animals generated high levels of Th17 cells-associated cytokines. Following disease induction, daily systemic administration of human recombinant Anx-A1 (hrAnx-A1), during the afferent phase of disease, restrained autoreactive CD4(+) cell proliferation, reduced expression of pro-inflammatory cytokines IL-17, IFN-γ and IL-6 and attenuated autoimmune retinal inflammatory disease. Furthermore, in man, Anx-A1 serum levels when measured in active uveitis patient sera were low and associated with the detection of IgM and IgG anti-Anx-A1 antibodies when compared to healthy individuals. This data supports Anx-A1 as an early and critical regulator of Th17 cell driven autoimmune diseases such as uveitis.


Asunto(s)
Anexina A1/administración & dosificación , Enfermedades Autoinmunes/inmunología , Proteínas Recombinantes/administración & dosificación , Células Th17/efectos de los fármacos , Uveítis/inmunología , Animales , Anexina A1/genética , Enfermedades Autoinmunes/inducido químicamente , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas del Ojo/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/inmunología , Proteínas Recombinantes/genética , Proteínas de Unión al Retinol/inmunología , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Células Th17/fisiología , Uveítis/inducido químicamente
5.
J Immunol ; 190(11): 5689-701, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23645879

RESUMEN

Annexin A1 (AnxA1) is a protein that displays potent anti-inflammatory properties, but its expression in eye tissue and its role in ocular inflammatory diseases have not been well studied. We investigated the mechanism of action and potential uses of AnxA1 and its mimetic peptide (Ac2-26) in the endotoxin-induced uveitis (EIU) rodent model and in human ARPE-19 cells activated by LPS. In rats, analysis of untreated EIU after 24 and 48 h or EIU treated with topical applications or with a single s.c. injection of Ac2-26 revealed the anti-inflammatory actions of Ac2-26 on leukocyte infiltration and on the release of inflammatory mediators; the systemic administration of Boc2, a formylated peptide receptor (fpr) antagonist, abrogated the peptide's protective effects. Moreover, AnxA1(-/-) mice exhibited exacerbated EIU compared with wild-type animals. Immunohistochemical studies of ocular tissue showed a specific AnxA1 posttranslational modification in EIU and indicated that the fpr2 receptor mediated the anti-inflammatory actions of AnxA1. In vitro studies confirmed the roles of AnxA1 and fpr2 and the protective effects of Ac2-26 on the release of chemical mediators in ARPE-19 cells. Molecular analysis of NF-κB translocation and IL-6, IL-8, and cyclooxygenase-2 gene expression indicated that the protective effects of AnxA1 occur independently of the NF-κB signaling pathway and possibly in a posttranscriptional manner. Together, our data highlight the role of AnxA1 in ocular inflammation, especially uveitis, and suggest the use of AnxA1 or its mimetic peptide Ac2-26 as a therapeutic approach.


Asunto(s)
Anexina A1/genética , Antiinflamatorios/farmacología , Péptidos/farmacología , Uveítis/genética , Animales , Anexina A1/administración & dosificación , Anexina A1/química , Anexina A1/metabolismo , Anexina A1/farmacología , Antiinflamatorios/administración & dosificación , Humor Acuoso/citología , Humor Acuoso/inmunología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Endotoxinas/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , FN-kappa B/metabolismo , Infiltración Neutrófila/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Oligopéptidos/farmacología , Péptidos/administración & dosificación , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Ratas , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Uveítis/inducido químicamente , Uveítis/inmunología
6.
Biomed Pharmacother ; 172: 116254, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38340398

RESUMEN

Leishmaniases, a group of diseases caused by the species of the protozoan parasite Leishmania, remains a significant public health concern worldwide. Host immune responses play a crucial role in the outcome of Leishmania infections, and several mediators that regulate inflammatory responses are potential targets for therapeutic approaches. Annexin A1 (AnxA1), an endogenous protein endowed with anti-inflammatory and pro-resolving properties, has emerged as a potential player. We have shown that during L. braziliensis infection, deficiency of AnxA1 exacerbates inflammatory responses but does not affect parasite burden. Here, we have investigated the role of AnxA1 in L. amazonensis infection, given the non-healing and progressive lesions characteristic of this infectious model. Infection of AnxA1 KO BALB/c mice resulted in increased lesion size and tissue damage associated with higher parasite burdens and enhanced inflammatory response. Notably, therapeutic application of the AnxA1 peptidomimetic Ac2-26 improves control of parasite replication and increases IL-10 production in vivo and in vitro, in both WT and AnxA1 KO mice. Conversely, administration of WRW4, an inhibitor of FPR2/3, resulted in larger lesions and decreased production of IL-10, suggesting that the effects of AnxA1 during L. amazonensis infection are associated with the engagement of these receptors. Our study illuminates the role of AnxA1 in L. amazonensis infection, demonstrating its impact on the susceptibility phenotype of BALB/c mice. Furthermore, our results indicate that targeting the AnxA1 pathway by using the Ac2-26 peptide could represent a promising alternative for new treatments for leishmaniasis.


Asunto(s)
Anexina A1 , Leishmania , Leishmaniasis , Péptidos , Animales , Ratones , Anexina A1/administración & dosificación , Anexina A1/metabolismo , Inmunidad , Interleucina-10/metabolismo , Leishmaniasis/tratamiento farmacológico , Ratones Endogámicos BALB C , Péptidos/administración & dosificación
7.
Biochem Biophys Res Commun ; 417(3): 1024-9, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22222376

RESUMEN

Chronic airway inflammation is a key feature of bronchial asthma. Annexin-1 (ANX1) is an anti-inflammatory protein that is an important modulator and plays a key role in inflammation. Although the precise action of ANX1 remains unclear, it has emerged as a potential drug target for inflammatory diseases such as asthma. To examine the protective effects of ANX1 protein on ovalbumin (OVA)-induced asthma in animal models, we used a cell-permeable Tat-ANX1 protein. Mice sensitized and challenged with OVA antigen had an increased amount of cytokines and eosinophils in their bronchoalveolar lavage (BAL) fluid. However, administration of Tat-ANX1 protein before OVA challenge significantly decreased the levels of cytokines (interleukin (IL)-4, IL-5, and IL-13) and BAL fluid in lung tissues. Furthermore, OVA significantly increased the activation of mitogen-activated protein kinase (MAPK) in lung tissues, whereas Tat-ANX1 protein markedly reduced phosphorylation of MAPKs such as extracellular signal-regulated protein kinase, p38, and stress-activated protein kinase/c-Jun N-terminal kinase. These results suggest that transduced Tat-ANX1 protein may be a potential protein therapeutic agent for the treatment of lung disorders including asthma.


Asunto(s)
Anexina A1/uso terapéutico , Anexinas/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Asma/tratamiento farmacológico , Productos del Gen tat/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Anexina A1/administración & dosificación , Anexinas/administración & dosificación , Antiinflamatorios no Esteroideos/administración & dosificación , Asma/prevención & control , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Productos del Gen tat/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ovalbúmina , Proteínas Recombinantes de Fusión/administración & dosificación
8.
Cells ; 10(11)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34831393

RESUMEN

Uveitis is one of the main causes of blindness worldwide, and therapeutic alternatives are worthy of study. We investigated the effects of piperlongumine (PL) and/or annexin A1 (AnxA1) mimetic peptide Ac2-26 on endotoxin-induced uveitis (EIU). Rats were inoculated with lipopolysaccharide (LPS) and intraperitoneally treated with Ac2-26 (200 µg), PL (200 and 400 µg), or Ac2-26 + PL after 15 min. Then, 24 h after LPS inoculation, leukocytes in aqueous humor, mononuclear cells, AnxA1, formyl peptide receptor (fpr)1, fpr2, and cyclooxygenase (COX)-2 were evaluated in the ocular tissues, along with inflammatory mediators in the blood and macerated supernatant. Decreased leukocyte influx, levels of inflammatory mediators, and COX-2 expression confirmed the anti-inflammatory actions of the peptide and pointed to the protective effects of PL at higher dosage. However, when PL and Ac2-26 were administered in combination, the inflammatory potential was lost. AnxA1 expression was elevated among groups treated with PL or Ac2-26 + PL but reduced after treatment with Ac2-26. Fpr2 expression was increased only in untreated EIU and Ac2-26 groups. The interaction between Ac2-26 and PL negatively affected the anti-inflammatory action of Ac2-26 or PL. We emphasize that the anti-inflammatory effects of PL can be used as a therapeutic strategy to protect against uveitis.


Asunto(s)
Anexina A1/uso terapéutico , Antiinflamatorios/uso terapéutico , Dioxolanos/uso terapéutico , Péptidos/uso terapéutico , Uveítis/inducido químicamente , Uveítis/tratamiento farmacológico , Animales , Anexina A1/administración & dosificación , Anexina A1/farmacología , Antiinflamatorios/farmacología , Cilios/enzimología , Cilios/patología , Ciclooxigenasa 2/metabolismo , Dioxolanos/administración & dosificación , Dioxolanos/farmacología , Endotoxinas , Ojo/efectos de los fármacos , Ojo/patología , Mediadores de Inflamación/metabolismo , Masculino , Modelos Biológicos , Monocitos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Péptidos/administración & dosificación , Péptidos/farmacología , Ratas Wistar , Receptores de Lipoxina/metabolismo , Uveítis/sangre , Uveítis/patología
9.
J Neuropathol Exp Neurol ; 66(10): 932-43, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17917587

RESUMEN

Annexin A1 (ANXA1) has been suggested to be a mediator of the anti-inflammatory actions of glucocorticoids and more recently an endogenous neuroprotective agent. In the present study, we investigated the anti-inflammatory and neuroprotective effects of ANXA1 in a model of contusive spinal cord injury (SCI). Here we report that injections of ANXA1 (Ac 2-26) into the acutely injured spinal cord at 2 concentrations (5 and 20 microg) inhibited SCI-induced increases in phospholipase A2 and myeloperoxidase activities. In addition, ANXA1 administration reduced the expression of interleukin-1beta and activated caspase-3 at 24 hours, and glial fibrillary acidic protein at 4 weeks postinjury. Furthermore, ANXA1 administration significantly reversed phospholipase A2-induced spinal cord neuronal death in vitro and reduced tissue damage and increased white matter sparing in vivo, compared to the vehicle-treated controls. Fluorogold retrograde tracing showed that ANXA1 administration protected axons of long descending pathways at 6 weeks post-SCI. ANXA1 administration also significantly increased the number of animals that responded to transcranial magnetic motor-evoked potentials. However, no measurable behavioral improvement was found after these treatments. These results, particularly the improvements obtained in tissue sparing and electrophysiologic measures, suggest a neuroprotective effect of ANXA1.


Asunto(s)
Anexina A1/uso terapéutico , Inflamación/prevención & control , Fosfolipasas A/metabolismo , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/enzimología , Animales , Anexina A1/administración & dosificación , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Electrofisiología , Activación Enzimática/efectos de los fármacos , Potenciales Evocados Motores/efectos de los fármacos , Potenciales Evocados Motores/fisiología , Femenino , Gliosis/patología , Inyecciones Espinales , Actividad Motora/efectos de los fármacos , Peroxidasa/metabolismo , Fosfolipasas A2 , Ratas , Ratas Sprague-Dawley , Médula Espinal/enzimología , Médula Espinal/patología , Traumatismos de la Médula Espinal/patología , Estilbamidinas
10.
J Clin Invest ; 125(3): 1215-27, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25664854

RESUMEN

Epithelial restitution is an essential process that is required to repair barrier function at mucosal surfaces following injury. Prolonged breaches in epithelial barrier function result in inflammation and further damage; therefore, a better understanding of the epithelial restitution process has potential for improving the development of therapeutics. In this work, we demonstrate that endogenous annexin A1 (ANXA1) is released as a component of extracellular vesicles (EVs) derived from intestinal epithelial cells, and these ANXA1-containing EVs activate wound repair circuits. Compared with healthy controls, patients with active inflammatory bowel disease had elevated levels of secreted ANXA1-containing EVs in sera, indicating that ANXA1-containing EVs are systemically distributed in response to the inflammatory process and could potentially serve as a biomarker of intestinal mucosal inflammation. Local intestinal delivery of an exogenous ANXA1 mimetic peptide (Ac2-26) encapsulated within targeted polymeric nanoparticles (Ac2-26 Col IV NPs) accelerated healing of murine colonic wounds after biopsy-induced injury. Moreover, one-time systemic administration of Ac2-26 Col IV NPs accelerated recovery following experimentally induced colitis. Together, our results suggest that local delivery of proresolving peptides encapsulated within nanoparticles may represent a potential therapeutic strategy for clinical situations characterized by chronic mucosal injury, such as is seen in patients with IBD.


Asunto(s)
Anexina A1/fisiología , Exosomas/fisiología , Mucosa Intestinal/fisiopatología , Animales , Anexina A1/administración & dosificación , Antiinflamatorios/administración & dosificación , Línea Celular , Colitis/sangre , Colitis/fisiopatología , Humanos , Mucosa Intestinal/efectos de los fármacos , Ratones Noqueados , Nanopartículas , Péptidos/administración & dosificación , Cicatrización de Heridas
11.
Carbohydr Polym ; 115: 629-35, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25439941

RESUMEN

In this paper, for the first time, hydrogels containing Annexin A1 N-terminal derived peptide, Ac2-26, as a novel dressing were successfully developed for dermal wound repair application. High mannuronic (M) content alginate and low molecular weight chitosan have been used as hydrogel carrier. Peptide recovery analyses, FTIR studies and molecular modelling highlighted chemical interactions between peptide and hydrogel polymers. Ac2-26 resulted entrapped into chitosan hydrogel matrix that prevented its release, whereas such interaction in alginate hydrogel slowed down peptide diffusion enabling its sustained release till 72 h. In vivo wound healing studies conducted on mice dorsal wounds indicate that after the 9th day of post wounding Ac2-26/alginate hydrogels could significantly accelerate wound healing, with complete closure of the wound on day 14th. Therefore, these results suggest that the developed of Ac2-26 high M content alginate hydrogel could be a promising wound dressing with potential application in dermal wound healing.


Asunto(s)
Anexina A1/administración & dosificación , Hidrogeles/administración & dosificación , Péptidos/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Administración Tópica , Alginatos/química , Animales , Anexina A1/química , Quitosano/química , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/química , Liberación de Fármacos , Hidrogeles/química , Inyecciones , Ratones Endogámicos C57BL , Péptidos/química
12.
PLoS One ; 10(6): e0130484, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26090792

RESUMEN

OBJECTIVE: To investigate therapeutic effects of annexin A1 (anxA1) on atherogenesis in LDLR-/- mice. METHODS: Human recombinant annexin A1 (hr-anxA1) was produced by a prokaryotic expression system, purified and analysed on phosphatidylserine (PS) binding and formyl peptide receptor (FPR) activation. Biodistribution of 99mTechnetium-hr-anxA1 was determined in C57Bl/6J mice. 12 Weeks old LDLR-/- mice were fed a Western Type Diet (WTD) during 6 weeks (Group I) or 12 weeks (Group P). Mice received hr-anxA1 (1 mg/kg) or vehicle by intraperitoneal injection 3 times per week for a period of 6 weeks starting at start of WTD (Group I) or 6 weeks after start of WTD (Group P). Total aortic plaque burden and phenotype were analyzed using immunohistochemistry. RESULTS: Hr-anxA1 bound PS in Ca2+-dependent manner and activated FPR2/ALX. It inhibited rolling and adherence of neutrophils but not monocytes on activated endothelial cells. Half lives of circulating 99mTc-hr-anxA1 were <10 minutes and approximately 6 hours for intravenously (IV) and intraperitoneally (IP) administered hr-anxA1, respectively. Pharmacological treatment with hr-anxA1 had no significant effect on initiation of plaque formation (-33%; P = 0.21)(Group I) but significantly attenuated progression of existing plaques of aortic arch and subclavian artery (plaque size -50%, P = 0.005; necrotic core size -76% P = 0.015, hr-anxA1 vs vehicle) (Group P). CONCLUSION: Hr-anxA1 may offer pharmacological means to treat chronic atherogenesis by reducing FPR-2 dependent neutrophil rolling and adhesion to activated endothelial cells and by reducing total plaque inflammation.


Asunto(s)
Anexina A1/farmacología , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Receptores de LDL/deficiencia , Proteínas Recombinantes , Animales , Anexina A1/administración & dosificación , Células Sanguíneas/metabolismo , Células de la Médula Ósea/metabolismo , Dieta Occidental/efectos adversos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Vías de Administración de Medicamentos , Humanos , Inmunofenotipificación , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/etiología , Receptores de LDL/genética
13.
Pain ; 109(1-2): 52-63, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15082126

RESUMEN

In this study we investigated how the peptides derived from the glucocorticoid-inducible protein annexin 1 are able to alter the nociceptive threshold of mice. The effects of the annexin1 fragment 2-26 (Anxa1(2-26)) on nociceptive threshold were studied using both chemical (formalin test) and thermal (hot plate and tail flick test) nociceptive stimuli on mice. Subcutaneous administration of Anxa1(2-26) into the dorsal surface of the mouse's hind paw was able to selectively reduce formalin-induced nociceptive behavior in the last phase of the test. The same effect was observed after intracerebroventricular administration, however, this was not the case when performing the hot plate or tail flick tests. Of the shortest Anxa1(2-26)-derived peptides, Anxa1(2-12) reduced the nociceptive response to formalin, however, the Anxa1(2-6) did not. The possible involvement of the receptors for formylated peptide in the anti-nociceptive action of Anxa1(-26) and Anxa1(2-12) was studied, choosing the formalin test. We found that the formyl peptide receptor agonist formyl-MLF (fMLF) induced anti-nociceptive effects in the formalin test both after the peripheral and central administration. The formyl peptide receptor antagonist N-t-butoxycarbonyl-MLP did not alter the response to formalin, but it was able to block the anti-nociceptive effects of Anxa1(2-26,) Anxa1(2-12) and fMLF after peripheral or central administration. These results indicate that exogenously administered Anxa1 can peripherally and centrally inhibit the nociceptive transmission associated with inflammatory processes through a mechanism that involves formyl peptide receptors.


Asunto(s)
Anexina A1/administración & dosificación , Dolor/tratamiento farmacológico , Receptores de Formil Péptido/fisiología , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Formaldehído , Calor , Inflamación/inducido químicamente , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inyecciones Intraventriculares , Masculino , Ratones , Dolor/etiología , Dimensión del Dolor/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Péptidos , Receptores de Formil Péptido/agonistas , Receptores de Formil Péptido/antagonistas & inhibidores , Factores de Tiempo
14.
J Neuroendocrinol ; 5(1): 51-61, 1993 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8485543

RESUMEN

Lipocortin 1 (LC1: also called annexin 1) was first described as a putative second messenger protein for the anti-inflammatory steroids in peripheral tissues. In the present study, in vitro and in vivo methods were used to examine its potential role within the hypothalamus as a mediator of the regulatory actions of the glucocorticoids on the hypothalamo-pituitary-adrenocortical axis of the rat. In the in vitro studies, the effects of human recombinant LC1 (hu-r-LC1) on the concomitant release of the two major corticotrophin-releasing factors (CRF-41 and arginine vasopressin, AVP) from isolated hypothalami removed from chronically adrenalectomized rats were compared with those of dexamethasone in the presence and absence of appropriate secretagogues, namely phospholipase A2 (PLA2), interleukin-6 (IL-6) and a non-specific depolarizing agent, K+ (56 mM). The spontaneous release of CRF-41 in vitro was unaffected by either hu-r-LC1 (5 to 100 ng/ml) or dexamethasone (1 microM). Both compounds however reduced the release of the neuropeptide evoked by IL-6 (5 ng/ml) but failed to modify the secretory responses to PLA2 (25 U/ml) or K+ (56 mM). Dexamethasone (1 microM) had no effect on the basal release of AVP but effectively blocked the secretion of the peptide induced by either IL-6 (10 ng/ml) or PLA2 (25 U/ml). In complete contrast, hu-r-LC1 (5 to 100 ng/ml) stimulated the release of AVP and potentiated the secretory responses to IL-6 (10 ng/ml) and PLA2 (25 U/ml) but not to K+ (56 mM). The hypothalamic responses to PLA2 stimulation (25 U/ml) were associated with significant (P < 0.01) increases in prostaglandin E2 release which, in some instances, were potentiated by hu-r-LC1 (5 to 20 ng/ml). In vivo, administration of histamine (0.6 mg/100 g body wt, ip) produced significant (P < 0.01) increases in the serum corticosterone concentration and in the hypothalamic LC1 content. Neither hu-r-LC1 (0.6 to 1.2 micrograms) nor a polyclonal anti-LC1 antibody (3 microliters, diluted 1:200), injected intracerebroventricularly (icv), influenced either the resting serum corticosterone concentration or the hypersecretion of the steroid evoked by histamine stress. A lower dose of the recombinant protein (0.3 micrograms icv) also failed to alter basal corticosterone release but, in contrast to the higher doses, potentiated the pituitary-adrenocortical responses to histamine. The results suggest that LC1 may contribute to some aspects of peptide release in the hypothalamus but that its actions are not necessarily related to those of the glucocorticoids.


Asunto(s)
Anexina A1/farmacología , Hormona Liberadora de Corticotropina/metabolismo , Dexametasona/farmacología , Adrenalectomía , Animales , Anexina A1/administración & dosificación , Anexina A1/inmunología , Anticuerpos Monoclonales/inmunología , Arginina Vasopresina/metabolismo , Dexametasona/administración & dosificación , Histamina/farmacología , Humanos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/fisiología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Técnicas In Vitro , Inyecciones Intraventriculares , Interleucina-6/farmacología , Masculino , Fosfolipasas A/farmacología , Fosfolipasas A2 , Neurohipófisis/efectos de los fármacos , Neurohipófisis/metabolismo , Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología
15.
Eur J Pharmacol ; 264(3): 379-84, 1994 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-7535228

RESUMEN

The anti-inflammatory action of nonapeptide fragments of uteroglobin or lipocortin I known as antiflammins, was tested in the carrageenan or phospholipase A2 rat paw oedema model. The development of carrageenan-induced oedema in rats was significantly inhibited during the early and late phases of the oedema by the local administration of antiflammins 1 and 2. However, the peptides were not able to inhibit phospholipase A2-induced oedema. The time course of the anti-oedematous activity of nonapeptides after intradermal carrageenan injection may be attributed to their effect on mast cell degranulation and accumulation and activation of leukocytes. Naja naja phospholipase A2 exhibited strong histamine release-inducing activity, which may have contributed to the rat paw oedema induction. Surprisingly, antiflammins had a limited but significant inhibitory effect on histamine secretion.


Asunto(s)
Anexina A1/farmacología , Ácidos Aristolóquicos , Edema/tratamiento farmacológico , Mastocitos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Uteroglobina/farmacología , Animales , Anexina A1/administración & dosificación , Anexina A1/uso terapéutico , Carragenina/administración & dosificación , Carragenina/toxicidad , Degranulación de la Célula/efectos de los fármacos , Clorfeniramina/uso terapéutico , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Edema/inducido químicamente , Venenos Elapídicos/enzimología , Elapidae , Liberación de Histamina/efectos de los fármacos , Indometacina/uso terapéutico , Inflamación/tratamiento farmacológico , Inyecciones Intradérmicas , Masculino , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/uso terapéutico , Fenantrenos/uso terapéutico , Fosfolipasas A/administración & dosificación , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A/toxicidad , Fosfolipasas A2 , Ratas , Ratas Sprague-Dawley , Uteroglobina/administración & dosificación , Uteroglobina/uso terapéutico
17.
J Immunol ; 179(12): 8533-43, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18056401

RESUMEN

The appropriate development of an inflammatory response is central for the ability of a host to deal with any infectious insult. However, excessive, misplaced, or uncontrolled inflammation may lead to acute or chronic diseases. The microbiota plays an important role in the control of inflammatory responsiveness. In this study, we investigated the role of lipoxin A4 and annexin-1 for the IL-10-dependent inflammatory hyporesponsiveness observed in germfree mice. Administration of a 15-epi-lipoxin A4 analog or an annexin-1-derived peptide to conventional mice prevented tissue injury, TNF-alpha production, and lethality after intestinal ischemia/reperfusion. This was associated with enhanced IL-10 production. Lipoxin A4 and annexin-1 failed to prevent reperfusion injury in IL-10-deficient mice. In germfree mice, there was enhanced expression of both lipoxin A4 and annexin-1. Blockade of lipoxin A4 synthesis with a 5-lipoxygenase inhibitor or Abs against annexin-1 partially prevented IL-10 production and this was accompanied by partial reversion of inflammatory hyporesponsiveness in germfree mice. Administration of BOC-1, an antagonist of ALX receptors (at which both lipoxin A4 and annexin-1 act), or simultaneous administration of 5-lipoxygenase inhibitor and anti-annexin-1 Abs, was associated with tissue injury, TNF-alpha production, and lethality similar to that found in conventional mice. Thus, our data demonstrate that inflammatory responsiveness is tightly controlled by the presence of the microbiota and that the innate capacity of germfree mice to produce IL-10 is secondary to their endogenous greater ability to produce lipoxin A4 and annexin-1.


Asunto(s)
Anexina A1/fisiología , Vida Libre de Gérmenes , Inflamación/inmunología , Interleucina-10/metabolismo , Lipoxinas/fisiología , Animales , Anexina A1/administración & dosificación , Anexina A1/antagonistas & inhibidores , Inflamación/prevención & control , Interleucina-10/genética , Intestinos , Lipoxinas/administración & dosificación , Lipoxinas/antagonistas & inhibidores , Ratones , Ratones Mutantes , Péptidos/administración & dosificación , Daño por Reperfusión/prevención & control
18.
Clin Exp Immunol ; 123(1): 62-7, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11167999

RESUMEN

Annexin 1 (ANX-1) can reduce leucocyte migration in response to cytokines and chemokines in some rodent models of inflammation. However, its effectiveness against an inflammatory stimulus as strong as bacterial lipopolysaccharide (LPS) is unknown. Thus, we have examined whether ANX-1 can modulate LPS-induced neutrophil accumulation in the rat, as assessed by intravital microscopy and by myeloperoxidase (MPO) assay. The anti-inflammatory glucocorticoid, dexamethasone (DEX) was also studied for comparison. LPS superfusion induced adhesion of leucocytes to the endothelium and a subsequent increase in emigration from rat post-capillary venules over 2 h as assessed by intravital microscopy. Either ANX-1 or DEX was able to attenuate this adhesion and emigration of leucocytes. MPO activity in the lung, kidney and ileum was elevated after a 6-h exposure to LPS (intraperitoneal), indicating accumulation of neutrophils in these tissues. DEX attenuated the LPS-induced increase in MPO in the ileum but had no effect on MPO in the lungs or kidneys. This would suggest that the underlying mechanism by which neutrophils accumulate in the ileum, and more generally in the gastrointestinal compartment, is different from other vascular beds. ANX-1 had no effect on the LPS-induced increase in MPO activity in any of the tissues studied. Thus, from these data, ANX-1 appears to reduce leucocyte adhesion and emigration induced by a short-term (2 h), but not a longer (6 h) exposure to LPS.


Asunto(s)
Anexina A1/farmacología , Dexametasona/farmacología , Lipopolisacáridos/administración & dosificación , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Neutrófilos/patología , Animales , Anexina A1/administración & dosificación , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Dexametasona/administración & dosificación , Endotoxemia/enzimología , Endotoxemia/inmunología , Endotoxemia/patología , Escherichia coli/inmunología , Técnicas In Vitro , Inyecciones Intravenosas , Inyecciones Subcutáneas , Masculino , Microscopía por Video , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Neutrófilos/inmunología , Perfusión , Peroxidasa/análisis , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA