Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Pathol ; 186(4): 962-73, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26878209

RESUMEN

Inflammation and its natural resolution are host-protective responses triggered by infection or injury. The resolution phase of inflammation is regulated by enzymatically produced specialized pro-resolving mediators. We recently identified a new class of peptide-conjugated specialized pro-resolving mediators that carry potent tissue regenerative actions that belong to the protectin family and are coined protectin conjugates in tissue regeneration (PCTR). Herein, with the use of microbial-induced peritonitis in mice and liquid chromatography-tandem mass spectrometry-based lipid mediator metabololipidomics, we found that PCTR1 is temporally regulated during self-resolving infection. When administered at peak of inflammation, PCTR1 enhanced macrophage recruitment and phagocytosis of Escherichia coli, decreased polymorphonuclear leukocyte infiltration, and counter-regulated inflammation-initiating lipid mediators, including prostaglandins. In addition, biologically produced PCTR1 promoted human monocyte and macrophage migration in a dose-dependent manner (0.001 to 10.0 nmol/L). We prepared PCTR1 via organic synthesis and confirmed that synthetic PCTR1 increased macrophage and monocyte migration, enhanced macrophage efferocytosis, and accelerated tissue regeneration in planaria. With human macrophage subsets, PCTR1 levels were significantly higher in M2 macrophages than in M1 phenotype, along with members of the resolvin conjugates in tissue regeneration and maresin conjugate families. In contrast, M1 macrophages gave higher levels of cysteinyl leukotrienes. Together, these results demonstrate that PCTR1 is a potent monocyte/macrophage agonist, regulating key anti-inflammatory and pro-resolving processes during bacterial infection.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Macrófagos/citología , Monocitos/efectos de los fármacos , Fagocitosis/fisiología , Animales , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacología , Escherichia coli/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Masculino , Ratones , Fagocitosis/efectos de los fármacos
2.
Eur J Immunol ; 46(1): 81-91, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26474728

RESUMEN

Specialized proresolving mediators (SPMs) constitute a recently recognized class of bioactive molecules thatpromote the resolution of inflammation. We recently reported that the SPMs resolvin D1 (RvD1) and 17-hydroxydocosahexaenoic acid (17-HDHA) promote the differentiation of IgG-secreting B cells and enhance antibody-mediated immune responses. However, there is an important knowledge gap regarding whether or not SPMs regulate human B-cell IgE production, which is the key effector in diseases such as asthma and allergy. Therefore, we investigated whether a panel of diverse SPMs influences B-cell IgE production. An important finding was that 17-HDHA and RvD1 inhibit IgE production by human B cells and suppress the differentiation of naïve B cells into IgE-secreting cells by specifically blocking epsilon germline transcript. This effect is specific to human IgE, as the SPMs do not inhibit production of IgM and IgG and did not suppress other IL-4-upregulated genes. 17-HDHA and RvD1 act by stabilizing the transcriptional repressor B-cell lymphoma 6, which competes with STAT6 for binding at the epsilon germline transcript promoter. Overall, these new findings demonstrate that certain SPMs inhibit the differentiation of IgE-producing B cells, without being broadly immune suppressive, representing a novel class of potential therapeutics for IgE-driven diseases such as asthma and allergy.


Asunto(s)
Linfocitos B/metabolismo , Ácidos Docosahexaenoicos/inmunología , Regulación de la Expresión Génica/inmunología , Inmunoglobulina E/biosíntesis , Activación de Linfocitos/inmunología , Linfocitos B/inmunología , Western Blotting , Diferenciación Celular/inmunología , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Cambio de Clase de Inmunoglobulina/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
FASEB J ; 29(5): 2120-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25713027

RESUMEN

Local mediators orchestrate the host response to both sterile and infectious challenge and resolution. Recent evidence demonstrates that maresin sulfido-conjugates actively resolve acute inflammation and promote tissue regeneration. In this report, we investigated self-limited infectious exudates for novel bioactive chemical signals in tissue regeneration and resolution. By use of spleens from Escherichia coli infected mice, self-resolving infectious exudates, human spleens, and blood from patients with sepsis, we identified 2 new families of potent molecules. Characterization of their physical properties and isotope tracking demonstrated that the bioactive structures contained a docosahexaenoate backbone and sulfido-conjugated triene or tetraene double-bond systems. Activated human phagocytes converted 17-hydro(peroxy)-4Z,7Z,10Z,13Z,15E,19Z-docosahexaenoic acid to these bioactive molecules. Regeneration of injured planaria was accelerated with nanomolar amounts of 16-glutathionyl, 17-hydroxy-4Z,7Z,10,12,14,19Z-docosahexaenoic acid and 16-cysteinylglycinyl, 17-hydroxy-4Z,7Z,10,12,14,19Z-docosahexaenoic acid (Protectin sulfido-conjugates) or 8-glutathionyl, 7,17-dihydroxy-4Z,9,11,13Z,15E,19Z-docosahexaenoic acid and 8-cysteinylglycinyl, 7,17-dihydroxy-4Z,9,11,13Z,15E,19Z-docosahexaenoic acid (Resolvin sulfido-conjugates). Each protectin and resolvin sulfido-conjugate dose dependently (0.1-10 nM) stimulated human macrophage bacterial phagocytosis, phagolysosomal acidification, and efferocytosis. Together, these results identify 2 novel pathways and provide evidence for structural elucidation of new resolution moduli. These resolvin and protectin conjugates identified in mice and human infected tissues control host responses promoting catabasis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antígenos CD59/química , Ácidos Docosahexaenoicos/química , Fagocitosis/efectos de los fármacos , Planarias/efectos de los fármacos , Regeneración/efectos de los fármacos , Animales , Células Cultivadas , Cromatografía Liquida , Escherichia coli/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Humanos , Inflamación/tratamiento farmacológico , Mediadores de Inflamación/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Peritonitis/tratamiento farmacológico , Peritonitis/microbiología , Peritonitis/patología , Planarias/citología , Regeneración/fisiología , Sepsis/tratamiento farmacológico , Sepsis/microbiología , Sepsis/patología , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Espectrometría de Masas en Tándem
4.
J Immunol ; 194(3): 863-7, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25539814

RESUMEN

Asthma is a chronic inflammatory disease that fails to resolve. Recently, a key role for type 2 innate lymphoid cells (ILC2s) was linked to asthma pathogenesis; however, mechanisms for ILC2 regulation remain to be determined. In this study, metabololipidomics of murine lungs identified temporal changes in endogenous maresin 1 (MaR1) during self-limited allergic inflammation. Exogenous MaR1 reduced lung inflammation and ILC2 expression of IL-5 and IL-13 and increased amphiregulin. MaR1 augmented de novo generation of regulatory T cells (Tregs), which interacted with ILC2s to markedly suppress cytokine production in a TGF-ß-dependent manner. Ab-mediated depletion of Tregs interrupted MaR1 control of ILC2 expression of IL-13 in vivo. Together, the findings uncover Tregs as potent regulators of ILC2 activation; MaR1 targets Tregs and ILC2s to restrain allergic lung inflammation, suggesting MaR1 as the basis for a new proresolving therapeutic approach to asthma and other chronic inflammatory diseases.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Inmunidad Innata , Activación de Linfocitos , Neumonía/inmunología , Neumonía/metabolismo , Subgrupos de Linfocitos T , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Inmunomodulación , Ratones , Ratones Transgénicos , Neumonía/terapia
5.
J Immunol ; 193(12): 6031-40, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25392529

RESUMEN

Influenza viruses remain a critical global health concern. More efficacious vaccines are needed to protect against influenza virus, yet few adjuvants are approved for routine use. Specialized proresolving mediators (SPMs) are powerful endogenous bioactive regulators of inflammation, with great clinical translational properties. In this study, we investigated the ability of the SPM 17-HDHA to enhance the adaptive immune response using an OVA immunization model and a preclinical influenza vaccination mouse model. Our findings revealed that mice immunized with OVA plus 17-HDHA or with H1N1-derived HA protein plus 17-HDHA increased Ag-specific Ab titers. 17-HDHA increased the number of Ab-secreting cells in vitro and the number of HA-specific Ab-secreting cells present in the bone marrow. Importantly, the 17-HDHA-mediated increased Ab production was more protective against live pH1N1 influenza infection in mice. To our knowledge, this is the first report on the biological effects of ω-3-derived SPMs on the humoral immune response. These findings illustrate a previously unknown biological link between proresolution signals and the adaptive immune system. Furthermore, this work has important implications for the understanding of B cell biology, as well as the development of new potential vaccine adjuvants.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Inmunidad Humoral/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/efectos de los fármacos , Formación de Anticuerpos/inmunología , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/efectos de los fármacos , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Masculino , Ratones , Infecciones por Orthomyxoviridae/virología , Células Plasmáticas/citología , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo
6.
J Nat Prod ; 77(10): 2241-7, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25247845

RESUMEN

Protectin D1 (PD1 (3)), a C22-dihydroxylated polyunsaturated fatty acid biosynthesized from all-Z-docosahexaenoic acid, belongs to the new family of endogenous mediators referred to as specialized pro-resolving lipid mediators. PD1 (3) is a natural product that displays potent anti-inflammatory properties together with pro-resolving actions including inhibition of polymorphonuclear leukocyte (PMN) infiltration and promotion of macrophage phagocytosis and efferocytosis. Given its potent endogenous actions, this compound has entered several clinical development programs. Little has been reported on the metabolism of PD1 (3). The synthesis and biological evaluations of the ω-22 monohydroxylated metabolite of PD1 (3), named herein 22-OH-PD1 (6), are presented. LC-MS/MS data of the free acid 6, obtained from hydrolysis of the synthetic methyl ester 7, matched data for the endogenously produced 22-OH-PD1 (6). Compound 6 exhibited potent pro-resolving actions by inhibiting PMN chemotaxis in vivo and in vitro comparable to its precursor PD1 (3) and decreased pro-inflammatory mediator levels in inflammatory exudates. The results reported herein provide new knowledge of the metabolism of the protectin class of specialized pro-resolving mediators.


Asunto(s)
Ácidos Docosahexaenoicos/síntesis química , Ácidos Docosahexaenoicos/farmacología , Antiinflamatorios/farmacología , Quimiotaxis/efectos de los fármacos , Ácidos Docosahexaenoicos/química , Ésteres , Ácidos Grasos Insaturados/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Estructura Molecular , Neutrófilos/efectos de los fármacos , Espectrometría de Masas en Tándem
7.
Eur J Immunol ; 44(2): 357-69, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24166736

RESUMEN

Specialized proresolving mediators are endogenous bioactive lipid molecules that play a fundamental role in the regulation of inflammation and its resolution. Lipoxins and other specialized proresolving mediators have been identified in important immunological tissues including bone marrow, spleen, and blood. Lipoxins regulate functions of the innate immune system including the promotion of monocyte recruitment and increase macrophage phagocytosis of apoptotic neutrophils. A major knowledge gap is whether lipoxins influence adaptive immune cells. Here, we analyzed the actions of lipoxin A4 (LXA4) and its receptor ALX/FPR2 on human and mouse B cells. LXA4 decreased IgM and IgG production on activated human B cells through ALX/FPR2-dependent signaling, which downregulated NF-κB p65 nuclear translocation. LXA4 also inhibited human memory B-cell antibody production and proliferation, but not naïve B-cell function. Lastly, LXA4 decreased antigen-specific antibody production in an OVA immunization mouse model. To our knowledge, this is the first description of the actions of lipoxins on human B cells, demonstrating a link between resolution signals and adaptive immunity. Regulating antibody production is crucial to prevent unwanted inflammation. Harnessing the ability of lipoxins to decrease memory B-cell antibody production can be beneficial to threat inflammatory and autoimmune disorders.


Asunto(s)
Inmunidad Adaptativa/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Linfocitos B/inmunología , Memoria Inmunológica/inmunología , Lipoxinas/inmunología , Receptores de Formil Péptido/inmunología , Receptores de Lipoxina/inmunología , Animales , Anticuerpos/inmunología , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo/inmunología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Inflamación/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas/inmunología , Transducción de Señal/inmunología , Regulación hacia Arriba/inmunología
8.
Curr Angiogenes ; 2(2): 135-142, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24883266

RESUMEN

Inflammation is implicated in the progression of multiple types of cancers including lung, colorectal, breast and hematological malignancies. Cyclooxygenases (Cox) -1 and -2 are important enzymes involved in the regulation of inflammation. Elevated Cox-2 expression is associated with a poor cancer prognosis. Hematological malignancies, which are among the top 10 most predominant cancers in the USA, express high levels of Cox-2. Current therapeutic approaches against hematological malignances are insufficient as many patients develop resistance or relapse. Therefore, targeting Cox-2 holds promise as a therapeutic approach to treat hematological malignancies. NSAIDs and Cox-2 selective inhibitors are anti-inflammatory drugs that decrease prostaglandin and thromboxane production while promoting the synthesis of specialized proresolving mediators. Here, we review the evidence regarding the applicability of NSAIDs, such as aspirin, as well as Cox-2 specific inhibitors, to treat hematological malignancies. Furthermore, we discuss how FDA-approved Cox inhibitors can be used as anti-cancer drugs alone or in combination with existing chemotherapeutic treatments.

9.
J Immunol ; 189(10): 4740-7, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23041568

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily. PPARγ, a ligand-activated transcription factor, has important anti-inflammatory and antiproliferative functions, and it has been associated with diseases including diabetes, scarring, and atherosclerosis, among others. PPARγ is expressed in most bone marrow-derived cells and influences their function. PPARγ ligands can stimulate human B cell differentiation and promote Ab production. A knowledge gap is that the role of PPARγ in B cells under physiological conditions is not known. We developed a new B cell-specific PPARγ (B-PPARγ) knockout mouse and explored the role of PPARγ during both the primary and secondary immune response. In this article, we show that PPARγ deficiency in B cells decreases germinal center B cells and plasma cell development, as well as the levels of circulating Ag-specific Abs during a primary challenge. Inability to generate germinal center B cells and plasma cells is correlated to decreased MHC class II expression and decreased Bcl-6 and Blimp-1 levels. Furthermore, B-PPARγ-deficient mice have an impaired memory response, characterized by low titers of Ag-specific Abs and low numbers of Ag-experienced, Ab-secreting cells. However, B-PPARγ-deficient mice have no differences in B cell population distribution within primary or secondary lymphoid organs during development. This is the first report, to our knowledge, to show that, under physiological conditions, PPARγ expression in B cells is required for an efficient B cell-mediated immune response as it regulates B cell differentiation and Ab production.


Asunto(s)
Anticuerpos/inmunología , Formación de Anticuerpos/fisiología , Especificidad de Anticuerpos/fisiología , Diferenciación Celular/inmunología , PPAR gamma/inmunología , Células Plasmáticas/inmunología , Animales , Anticuerpos/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Humanos , Ratones , Ratones Noqueados , Especificidad de Órganos , PPAR gamma/genética , Células Plasmáticas/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-bcl-6 , Factores de Transcripción/genética , Factores de Transcripción/inmunología
10.
PPAR Res ; 2012: 705352, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22778711

RESUMEN

Peroxisome proliferator activated receptor (PPAR)-γ is a nuclear hormone receptor that is activated by multiple agonists including thiazolidinediones, prostaglandins, and synthetic oleanolic acids. Many PPARγ ligands are under investigation as potential therapies for human diseases. These ligands modulate multiple cellular pathways via both PPARγ-dependent and PPARγ-independent mechanisms. Here, we review the role of PPARγ and PPARγ ligands in lung disease, with emphasis on PPARγ-independent effects. PPARγ ligands show great promise in moderating lung inflammation, as antiproliferative agents in combination to enhance standard chemotherapy in lung cancer and as treatments for pulmonary fibrosis, a progressive fatal disease with no effective therapy. Some of these effects occur when PPARγ is pharmaceutically antagonized or genetically PPARγ and are thus independent of classical PPARγ-dependent transcriptional control. Many PPARγ ligands demonstrate direct binding to transcription factors and other proteins, altering their function and contributing to PPARγ-independent inhibition of disease phenotypes. These PPARγ-independent mechanisms are of significant interest because they suggest new therapeutic uses for currently approved drugs and because they can be used as probes to identify novel proteins and pathways involved in the pathogenesis or treatment of disease, which can then be targeted for further investigation and drug development.

11.
J Immunol ; 189(2): 1036-42, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22711890

RESUMEN

The resolution of inflammation is an active and dynamic process critical in maintaining homeostasis. Newly identified lipid mediators have been recognized as key players during the resolution phase. These specialized proresolving mediators (SPM) constitute separate families that include lipoxins, resolvins, protectins, and maresins, each derived from essential polyunsaturated fatty acids. New results demonstrate that SPM regulate aspects of the immune response, including reduction of neutrophil infiltration, decreased T cell cytokine production, and stimulation of macrophage phagocytic activity. The actions of SPM on B lymphocytes remain unknown. Our study shows that the novel SPM 17-hydroxydosahexaenoic acid (17-HDHA), resolvin D1, and protectin D1 are present in the spleen. Interestingly, 17-HDHA and resolvin D1, but not protectin D1, strongly increase activated human B cell IgM and IgG production. Furthermore, increased Ab production by 17-HDHA is due to augmented B cell differentiation toward a CD27(+)CD38(+) Ab-secreting cell phenotype. The 17-HDHA did not affect proliferation and was nontoxic to cells. Increase of plasma cell differentiation and Ab production supports the involvement of SPM during the late stages of inflammation and pathogen clearance. The present study provides new evidence for SPM activity in the humoral response. These new findings highlight the potential applications of SPM as endogenous and nontoxic adjuvants, and as anti-inflammatory therapeutic molecules.


Asunto(s)
Células Productoras de Anticuerpos/citología , Células Productoras de Anticuerpos/inmunología , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/inmunología , Diferenciación Celular/inmunología , Ácidos Docosahexaenoicos/fisiología , Regulación hacia Arriba/inmunología , Animales , Células Productoras de Anticuerpos/metabolismo , Subgrupos de Linfocitos B/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Liquida , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Metabolómica/métodos , Ratones , Ratones Endogámicos , Espectrometría de Masas en Tándem , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...