Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Signal ; 17(824): eadg9256, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38377179

RESUMO

High-density lipoprotein (HDL) nanoparticles promote endothelial cell (EC) function and suppress inflammation, but their utility in treating EC dysfunction has not been fully explored. Here, we describe a fusion protein named ApoA1-ApoM (A1M) consisting of apolipoprotein A1 (ApoA1), the principal structural protein of HDL that forms lipid nanoparticles, and ApoM, a chaperone for the bioactive lipid sphingosine 1-phosphate (S1P). A1M forms HDL-like particles, binds to S1P, and is signaling competent. Molecular dynamics simulations showed that the S1P-bound ApoM moiety in A1M efficiently activated EC surface receptors. Treatment of human umbilical vein ECs with A1M-S1P stimulated barrier function either alone or cooperatively with other barrier-enhancing molecules, including the stable prostacyclin analog iloprost, and suppressed cytokine-induced inflammation. A1M-S1P injection into mice during sterile inflammation suppressed neutrophil influx and inflammatory mediator secretion. Moreover, systemic A1M administration led to a sustained increase in circulating HDL-bound S1P and suppressed inflammation in a murine model of LPS-induced endotoxemia. We propose that A1M administration may enhance vascular endothelial barrier function, suppress cytokine storm, and promote resilience of the vascular endothelium.


Assuntos
Apolipoproteínas , Lipocalinas , Humanos , Camundongos , Animais , Apolipoproteínas/metabolismo , Apolipoproteínas/farmacologia , Lipocalinas/metabolismo , Lipocalinas/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Apolipoproteínas M , Inflamação , Lipoproteínas HDL/farmacologia , Lipoproteínas HDL/metabolismo , Lisofosfolipídeos/farmacologia , Lisofosfolipídeos/metabolismo , Esfingosina
2.
Autophagy ; 19(8): 2296-2317, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36781380

RESUMO

LCN2/neutrophil gelatinase-associated lipocalin/24p3 (lipocalin 2) is a secretory protein that acts as a mammalian bacteriostatic molecule. Under neuroinflammatory stress conditions, LCN2 is produced and secreted by activated microglia and reactive astrocytes, resulting in neuronal apoptosis. However, it remains largely unknown whether inflammatory stress and neuronal loss can be minimized by modulating LCN2 production and secretion. Here, we first demonstrated that LCN2 was secreted from reactive astrocytes, which were stimulated by treatment with lipopolysaccharide (LPS) as an inflammatory stressor. Notably, we found two effective conditions that led to the reduction of induced LCN2 levels in reactive astrocytes: proteasome inhibition and macroautophagic/autophagic flux activation. Mechanistically, proteasome inhibition suppresses NFKB/NF-κB activation through NFKBIA/IκBα stabilization in primary astrocytes, even under inflammatory stress conditions, resulting in the downregulation of Lcn2 expression. In contrast, autophagic flux activation via MTOR inhibition reduced the intracellular levels of LCN2 through its pre-secretory degradation. In addition, we demonstrated that the N-terminal signal peptide of LCN2 is critical for its secretion and degradation, suggesting that these two pathways may be mechanistically coupled. Finally, we observed that LPS-induced and secreted LCN2 levels were reduced in the astrocyte-cultured medium under the above-mentioned conditions, resulting in increased neuronal viability, even under inflammatory stress.Abbreviations: ACM, astrocyte-conditioned medium; ALP, autophagy-lysosome pathway; BAF, bafilomycin A1; BTZ, bortezomib; CHX, cycloheximide; CNS, central nervous system; ER, endoplasmic reticulum; GFAP, glial fibrillary acidic protein; GFP, green fluorescent protein; JAK, Janus kinase; KD, knockdown; LCN2, lipocalin 2; LPS, lipopolysaccharide; MACS, magnetic-activated cell sorting; MAP1LC3/LC3, microtubule-associated protein 1 light chain 3; MTOR, mechanistic target of rapamycin kinase; NFKB/NF-κB, nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105; NFKBIA/IκBα, nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha; OVEX, overexpression; SLC22A17, solute carrier family 22 member 17; SP, signal peptide; SQSTM1, sequestosome 1; STAT3, signal transducer and activator of transcription 3; TNF/TNF-α, tumor necrosis factor; TUBA, tubulin, alpha; TUBB3/ß3-TUB, tubulin, beta 3 class III; UB, ubiquitin; UPS, ubiquitin-proteasome system.


Assuntos
Lipocalinas , NF-kappa B , Animais , Lipocalinas/genética , Lipocalinas/metabolismo , Lipocalinas/farmacologia , Lipocalina-2/metabolismo , Lipocalina-2/farmacologia , NF-kappa B/metabolismo , Astrócitos/metabolismo , Tubulina (Proteína)/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , Inibidor de NF-kappaB alfa/farmacologia , Lipopolissacarídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Autofagia , Sistema Nervoso Central/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Mamíferos/metabolismo
3.
Brain Res Bull ; 189: 80-101, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-35988785

RESUMO

Astrocytes are the main support cells of the central nervous system. They also participate in neuroimmune reactions. In response to pathological and immune stimuli, astrocytes transform to reactive states characterized by increased release of inflammatory mediators. Some of these molecules are neuroprotective and inflammation resolving while others, including reactive oxygen species (ROS), nitric oxide (NO), matrix metalloproteinase (MMP)- 9, L-glutamate, and tumor necrosis factor α (TNF), are well-established toxins known to cause damage to surrounding cells and tissues. We hypothesized that similar to microglia, the brain immune cells, reactive astrocytes can release a broader set of diverse molecules that are potentially neurotoxic. A literature search was conducted to identify such molecules using the following two criteria: 1) evidence of their expression and secretion by astrocytes and 2) direct neurotoxic action. This review describes 14 structurally diverse molecules as less-established astrocyte neurotoxins, including C-X-C motif chemokine ligand (CXCL)10, CXCL12/CXCL12(5-67), FS-7-associated surface antigen ligand (FasL), macrophage inflammatory protein (MIP)- 2α, TNF-related apoptosis inducing ligand (TRAIL), pro-nerve growth factor (proNGF), pro-brain-derived neurotrophic factor (proBDNF), chondroitin sulfate proteoglycans (CSPGs), cathepsin (Cat)B, group IIA secretory phospholipase A2 (sPLA2-IIA), amyloid beta peptides (Aß), high mobility group box (HMGB)1, ceramides, and lipocalin (LCN)2. For some of these molecules, further studies are required to establish either their direct neurotoxic effects or the full spectrum of stimuli that induce their release by astrocytes. Only limited studies with human-derived astrocytes and neurons are available for most of these potential neurotoxins, which is a knowledge gap that should be addressed in the future. We also summarize available evidence of the role these molecules play in select neuropathologies where reactive astrocytes are a key feature. A comprehensive understanding of the full spectrum of neurotoxins released by reactive astrocytes is key to understanding neuroinflammatory diseases characterized by the adverse activation of these cells and may guide the development of novel treatment strategies.


Assuntos
Síndromes Neurotóxicas , Fosfolipases A2 Secretórias , Peptídeos beta-Amiloides/metabolismo , Antígenos de Superfície/metabolismo , Antígenos de Superfície/farmacologia , Astrócitos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Catepsinas/metabolismo , Ceramidas , Quimiocinas/metabolismo , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Ácido Glutâmico/metabolismo , Proteínas HMGB/metabolismo , Proteínas HMGB/farmacologia , Humanos , Mediadores da Inflamação/metabolismo , Ligantes , Lipocalinas/metabolismo , Lipocalinas/farmacologia , Proteínas Inflamatórias de Macrófagos/metabolismo , Proteínas Inflamatórias de Macrófagos/farmacologia , Microglia/metabolismo , Síndromes Neurotóxicas/metabolismo , Neurotoxinas/toxicidade , Óxido Nítrico/metabolismo , Fosfolipases A2 Secretórias/metabolismo , Fosfolipases A2 Secretórias/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
Cell Death Differ ; 29(3): 642-656, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34743203

RESUMO

Glycoprotein prostaglandin D2 synthase (PTGDS) is a member of the lipocalin superfamily and plays dual roles in prostaglandins metabolism and lipid transport. PTGDS has been involved in various cellular processes including the tumorigenesis of solid tumors, yet its role in carcinogenesis is contradictory and the significance of PTGDS in hematological malignancies is ill-defined. Here, we aimed to explore the expression and function of PTGDS in diffuse large B-cell lymphoma (DLBCL), especially the potential role of PTGDS inhibitor, AT56, in lymphoma therapy. Remarkable high expression of PTGDS was found in DLBCL, which was significantly correlated with poor prognosis. PTGDS overexpression and rhPTGDS were found to promote cell proliferation. Besides, in vitro and in vivo studies indicated that PTGDS knockdown and AT56 treatment exerted an anti-tumor effect by regulating cell viability, proliferation, apoptosis, cell cycle, and invasion, and enhanced the drug sensitivity to adriamycin and bendamustine through promoting DNA damage. Moreover, the co-immunoprecipitation-based mass spectrum identified the interaction between PTGDS and MYH9, which was found to promote DLBCL progression. PTGDS inhibition led to reduced expression of MYH9, and then declined activation of the Wnt-ß-catenin-STAT3 pathway through influencing the ubiquitination and degradation of GSK3-ß in DLBCL. The rescue experiment demonstrated that PTGDS exerted an oncogenic role through regulating MYH9 and then the Wnt-ß-catenin-STAT3 pathway. Based on point mutation of glycosylation sites, we confirmed the N-glycosylation of PTGDS in Asn51 and Asn78 and found that abnormal glycosylation of PTGDS resulted in its nuclear translocation, prolonged half-life, and enhanced cell proliferation. Collectively, our findings identified for the first time that glycoprotein PTGDS promoted tumorigenesis of DLBCL through MYH9-mediated regulation of Wnt-ß-catenin-STAT3 signaling, and highlighted the potential role of AT56 as a novel therapeutic strategy for DLBCL treatment.


Assuntos
Oxirredutases Intramoleculares/metabolismo , Linfoma Difuso de Grandes Células B , beta Catenina , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Lipocalinas/genética , Lipocalinas/metabolismo , Lipocalinas/farmacologia , Linfoma Difuso de Grandes Células B/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/metabolismo
5.
PLoS One ; 14(3): e0212023, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30917125

RESUMO

In chronic kidney disease both renal insufficiency and chronic inflammation trigger elevated hepcidin levels, which impairs iron uptake, availability. and erythropoiesis. Here we report the two first-in-human phase 1 trials of PRS-080#22, a novel, rationally engineered Anticalin protein that targets and antagonizes hepcidin. A single intravenous infusion of placebo or PRS-080#22 was administered to 48 healthy volunteers (phase 1a) and 24 patients with end stage chronic kidney disease (CKD) on hemodialysis (phase 1b) at different doses (0.08-16mg/kg for the phase 1a study and 2-8mg/kg for the phase 1b study) in successive dosing cohorts. The primary endpoint for both randomized, double-blind, phase 1 trials was safety and tolerability. Following treatment, all subjects were evaluable, with none experiencing dose limiting toxicities. Most adverse events were mild. One serious adverse event occurred in the phase 1b (CKD patient) study. There were no clinically significant changes in safety laboratory values or vital signs. PRS-080#22 showed dose-proportional pharmacokinetics (PK), with a terminal half-life of approximately three days in healthy volunteers and 10 to 12 days in CKD patients. Serum hepcidin levels were suppressed in a dose dependent manner and remained low for up to 48 hours after dosing. PRS-080#22 dose-dependently mobilized serum iron with increases in both serum iron concentration and transferrin saturation. No consistent changes were observed with regard to ferritin, reticulocytes, hemoglobin, and reticulocyte hemoglobin. Low titer anti-drug-antibodies were detected in five healthy volunteers but in none of the CKD patients. PRS-080#22, a novel Anticalin protein with picomolar affinity for hepcidin, was safe and well-tolerated when administered to healthy volunteers and CKD patients at all doses tested. The drug exhibited linear pharmacokinetics, longer half-life in CKD patients in comparison to healthy volunteers as well as expected pharmacodynamic effects which hold promise for further clinical studies.


Assuntos
Hepcidinas/antagonistas & inibidores , Lipocalinas/farmacologia , Insuficiência Renal Crônica/tratamento farmacológico , Adulto , Método Duplo-Cego , Feminino , Meia-Vida , Voluntários Saudáveis , Humanos , Infusões Intravenosas , Lipocalinas/farmacocinética , Masculino , Pessoa de Meia-Idade , Diálise Renal/métodos
6.
PLoS Genet ; 13(2): e1006603, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28182653

RESUMO

Environmental insults such as oxidative stress can damage cell membranes. Lysosomes are particularly sensitive to membrane permeabilization since their function depends on intraluminal acidic pH and requires stable membrane-dependent proton gradients. Among the catalog of oxidative stress-responsive genes is the Lipocalin Apolipoprotein D (ApoD), an extracellular lipid binding protein endowed with antioxidant capacity. Within the nervous system, cell types in the defense frontline, such as astrocytes, secrete ApoD to help neurons cope with the challenge. The protecting role of ApoD is known from cellular to organism level, and many of its downstream effects, including optimization of autophagy upon neurodegeneration, have been described. However, we still cannot assign a cellular mechanism to ApoD gene that explains how this protection is accomplished. Here we perform a comprehensive analysis of ApoD intracellular traffic and demonstrate its role in lysosomal pH homeostasis upon paraquat-induced oxidative stress. By combining single-lysosome in vivo pH measurements with immunodetection, we demonstrate that ApoD is endocytosed and targeted to a subset of vulnerable lysosomes in a stress-dependent manner. ApoD is functionally stable in this acidic environment, and its presence is sufficient and necessary for lysosomes to recover from oxidation-induced alkalinization, both in astrocytes and neurons. This function is accomplished by preventing lysosomal membrane permeabilization. Two lysosomal-dependent biological processes, myelin phagocytosis by astrocytes and optimization of neurodegeneration-triggered autophagy in a Drosophila in vivo model, require ApoD-related Lipocalins. Our results uncover a previously unknown biological function of ApoD, member of the finely regulated and evolutionary conserved gene family of extracellular Lipocalins. They set a lipoprotein-mediated regulation of lysosomal membrane integrity as a new mechanism at the hub of many cellular functions, critical for the outcome of a wide variety of neurodegenerative diseases. These results open therapeutic opportunities by providing a route of entry and a repair mechanism for lysosomes in pathological situations.


Assuntos
Astrócitos/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Apolipoproteínas D/genética , Apolipoproteínas D/metabolismo , Apolipoproteínas D/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/ultraestrutura , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular Tumoral , Células Cultivadas , Drosophila , Células HEK293 , Herbicidas/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Immunoblotting , Lipocalinas/farmacologia , Lisossomos/química , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Modelos Biológicos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/prevenção & controle , Neurônios/efeitos dos fármacos , Paraquat/farmacologia , Fagossomos/metabolismo
7.
Lipids Health Dis ; 16(1): 36, 2017 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-28179022

RESUMO

BACKGROUND: High-density Lipoprotein (HDL) attenuates endothelial cell apoptosis induced by different cell-death stimuli such as oxidation or growth factor deprivation. HDL is the main plasma carrier of the bioactive lipid sphingosine 1-phosphate (S1P), which it is a signaling molecule that promotes cell survival in response to several apoptotic stimuli. In HDL, S1P is bound to Apolipoprotein M (ApoM), a Lipocalin that is only present in around 5% of the HDL particles. The goal of this study is to characterize ApoM-bound S1P role in endothelial apoptosis protection and the signaling pathways involved. METHODS: Human umbilical vein endothelial cells (HUVEC) cultures were switched to serum/grow factor deprivation medium to induce apoptosis and the effect caused by the addition of ApoM and S1P analyzed. RESULTS: The addition of HDL+ApoM or recombinant ApoM-bound S1P promoted cell viability and blocked apoptosis, whereas HDL-ApoM had no protective effect. Remarkably, S1P exerted a more potent anti-apoptotic effect when carried by ApoM as compared to albumin, or when added as free molecule. Mechanistically, cooperation between S1P1 and S1P3 was required for the HDL/ApoM/S1P-mediated anti-apoptotic ability. Furthermore, AKT and ERK phosphorylation was also necessary to achieve the anti-apoptotic effect of the HDL/ApoM/S1P complex. CONCLUSIONS: Altogether, our results indicate that ApoM and S1P are key elements of the anti-apoptotic activity of HDL and promote optimal endothelial function.


Assuntos
Apolipoproteínas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Lipocalinas/metabolismo , Lipoproteínas HDL/metabolismo , Lisofosfolipídeos/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Apolipoproteínas/genética , Apolipoproteínas/farmacologia , Apolipoproteínas M , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultura/química , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Lipocalinas/genética , Lipocalinas/farmacologia , Lipoproteínas HDL/genética , Lipoproteínas HDL/farmacologia , Lisofosfolipídeos/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Lisoesfingolipídeo/genética , Albumina Sérica/farmacologia , Esfingosina/metabolismo , Esfingosina/farmacologia
8.
Arterioscler Thromb Vasc Biol ; 37(1): 118-129, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27879252

RESUMO

OBJECTIVE: Plasma high-density lipoproteins (HDL) are potent antiatherogenic and anti-inflammatory particles. However, HDL particles are highly heterogenic in composition, and different HDL-mediated functions can be ascribed to different subclasses of HDL. Only a small HDL population contains apolipoprotein M (ApoM), which is the main plasma carrier of the bioactive lipid mediator sphingosine-1-phosphate (S1P). Vascular inflammation is modulated by S1P, but both pro- and anti-inflammatory roles have been ascribed to S1P. The goal of this study is to elucidate the role of ApoM and S1P in endothelial anti-inflammatory events related to HDL. APPROACH AND RESULTS: Aortic or brain human primary endothelial cells were challenged with tumor necrosis factor-α (TNF-α) as inflammatory stimuli. The presence of recombinant ApoM-bound S1P or ApoM-containing HDL reduced the abundance of adhesion molecules in the cell surface, whereas ApoM and ApoM-lacking HDL did not. Specifically, ApoM-bound S1P decreased vascular adhesion molecule-1 (VCAM-1) and E-selectin surface abundance but not intercellular adhesion molecule-1. Albumin, which is an alternative S1P carrier, was less efficient in inhibiting VCAM-1 than ApoM-bound S1P. The activation of the S1P receptor 1 was sufficient and required to promote anti-inflammation. Moreover, ApoM-bound S1P induced the rearrangement of the expression of S1P-related genes to counteract TNF-α. Functionally, HDL/ApoM/S1P limited monocyte adhesion to the endothelium and maintained endothelial barrier integrity under inflammatory conditions. CONCLUSIONS: ApoM-bound S1P is a key component of HDL and is responsible for several HDL-associated protective functions in the endothelium, including regulation of adhesion molecule abundance, leukocyte-endothelial adhesion, and endothelial barrier.


Assuntos
Anti-Inflamatórios/farmacologia , Apolipoproteínas/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Inflamação/prevenção & controle , Lipocalinas/farmacologia , Lisofosfolipídeos/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Esfingosina/análogos & derivados , Anti-Inflamatórios/metabolismo , Apolipoproteínas/metabolismo , Apolipoproteínas M , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Selectina E/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Lipocalinas/metabolismo , Lipoproteínas HDL/metabolismo , Lisofosfolipídeos/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Ligação Proteica , Receptores de Lisoesfingolipídeo/metabolismo , Proteínas Recombinantes/farmacologia , Albumina Sérica/metabolismo , Albumina Sérica/farmacologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/metabolismo , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Fatores de Tempo , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
9.
J Biomed Mater Res A ; 105(3): 847-858, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27885823

RESUMO

For targeted brain delivery, nanoparticles (NPs) should bypass the blood-brain barrier (BBB). Novel functionalization strategies, based on low-density lipoprotein receptor (LDLR) binding domain, have been here tested to increase the brain targeting efficacy of poly d,l-lactic-co-glycolic acid (PLGA) NPs, biodegradable and suited for biomedical applications. Custom-made PLGA NPs were functionalized with an apolipoprotein E modified peptide (pep-apoE) responsible for LDLR binding, or with lipocalin-type prostaglandin-d-synthase (L-PGDS), highly expressed in the brain. At the comparison of pep-apoE and L-PGDS sequences, a highly homologs region was here identified, indicating that also L-PGDS could bind LDLR. Non-functionalized and functionalized NPs did not affect the viability of cultured human dendritic cells, protagonists of the immune response, and did not activate them to a proinflammatory profile. At 2 h after intravenous injection in mice, functionalized, but not the non-functionalized ones, fluorescent-tagged NPs were observed in the cerebral cortex parenchyma. The NPs were mostly internalized by neurons and microglia; glial cells showed a weak activation. The findings indicate that the tested functionalization strategies do not elicit adverse immune responses and that the peptidic moieties enable BBB traversal of the NPs, thus providing potential brain drug carriers. These could be especially effective for brain diseases in which LDLR is involved. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 847-858, 2017.


Assuntos
Barreira Hematoencefálica/metabolismo , Córtex Cerebral/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Oxirredutases Intramoleculares , Ácido Láctico , Lipocalinas , Nanopartículas , Peptídeos , Ácido Poliglicólico , Receptores de LDL/química , Apolipoproteínas E/química , Apolipoproteínas E/farmacocinética , Apolipoproteínas E/farmacologia , Feminino , Humanos , Oxirredutases Intramoleculares/química , Oxirredutases Intramoleculares/farmacocinética , Oxirredutases Intramoleculares/farmacologia , Ácido Láctico/química , Ácido Láctico/farmacocinética , Ácido Láctico/farmacologia , Lipocalinas/química , Lipocalinas/farmacocinética , Lipocalinas/farmacologia , Masculino , Nanopartículas/química , Nanopartículas/uso terapêutico , Peptídeos/química , Peptídeos/farmacocinética , Peptídeos/farmacologia , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacocinética , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
10.
Biochem J ; 473(11): 1563-78, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27029347

RESUMO

Amyloid beta (Aß) peptides, in particular Aß42 and Aß40, exert neurotoxic effects and their overproduction leads to amyloid deposits in the brain, thus constituting an important biomolecular target for treatments of Alzheimer's disease (AD). We describe the engineering of cognate Anticalins as a novel type of neutralizing protein reagent based on the human lipocalin scaffold. Phage display selection from a genetic random library comprising variants of the human lipocalin 2 (Lcn2) with mutations targeted at 20 exposed amino acid positions in the four loops that form the natural binding site was performed using both recombinant and synthetic target peptides and resulted in three different Anticalins. Biochemical characterization of the purified proteins produced by periplasmic secretion in Escherichia coli revealed high folding stability in a monomeric state, with Tm values ranging from 53.4°C to 74.5°C, as well as high affinities for Aß40, between 95 pM and 563 pM, as measured by real-time surface plasmon resonance analysis. The central linear VFFAED epitope within the Aß sequence was mapped using a synthetic peptide array on membranes and was shared by all three Anticalins, despite up to 13 mutual amino acid differences in their binding sites. All Anticalins had the ability-with varying extent-to inhibit Aß aggregation in vitro according to the thioflavin-T fluorescence assay and, furthermore, they abolished Aß42-mediated toxicity in neuronal cell culture. Thus, these Anticalins provide not only useful protein reagents to study the molecular pathology of AD but they also show potential as alternative drug candidates compared with antibodies.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Lipocalinas/química , Engenharia de Proteínas/métodos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Lipocalinas/farmacologia , Lipocalinas/uso terapêutico , Dados de Sequência Molecular , Biblioteca de Peptídeos , Ligação Proteica , Conformação Proteica
11.
J Gastroenterol Hepatol ; 31(1): 145-54, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26189649

RESUMO

BACKGROUND AND AIM: Because neutrophil gelatinase-associated lipocalin (NGAL) is known to provide significant bacteriostatic effects during infectious conditions, we tested the hypothesis that this protein is up-regulated and secreted into the intraluminal cavity of the gut under critically ill conditions and is thus responsible for the regulation of bacterial overgrowth. METHODS: With our institutional approval, male C57BL/6J mouse (6-7 weeks) were enrolled and applied for lipopolysaccharide or peritonitis model compared with naïve control. We assessed NGAL protein concentrations in intestinal lumen and up-regulation of NGAL expression in intestinal tissues in in vivo as well as ex vivo settings. Simultaneously, we examined the effects of NGAL protein administration on the growth of Escherichia coli (E. coli) in in vivo and in vitro experimental settings. The localization of NGAL in intestinal tissues and lumen was also assessed by immunohistological approach using NGAL antibody. RESULTS: Both lipopolysaccharide and peritonitis insults evoked the marked up-regulation of NGAL mRNA and protein levels in gut tissues such as crypt cells. In addition, the administration of NGAL protein significantly inhibited the outgrowth of enteric E. coli under both in vitro and in vivo conditions, accompanied by histological evidence. CONCLUSION: Neutrophil gelatinase-associated lipocalin protein accompanied by apparent bacteriostatic action accumulated in the intestinal wall and streamed into the mucosal layer during critically ill state, thereby possibly shaping microbiota homeostasis in the gut.


Assuntos
Proteínas de Fase Aguda/farmacologia , Proteínas de Fase Aguda/fisiologia , Intestinos/microbiologia , Lipocalinas/farmacologia , Lipocalinas/fisiologia , Microbiota/efeitos dos fármacos , Proteínas Oncogênicas/farmacologia , Proteínas Oncogênicas/fisiologia , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Animais , Estado Terminal , Modelos Animais de Doenças , Escherichia coli/crescimento & desenvolvimento , Expressão Gênica , Homeostase/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Lipocalina-2 , Lipocalinas/genética , Lipocalinas/metabolismo , Lipopolissacarídeos , Masculino , Camundongos Endogâmicos C57BL , Microbiota/fisiologia , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Peritonite/microbiologia , Regulação para Cima
12.
Angiogenesis ; 19(1): 79-94, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26650228

RESUMO

Human tear lipocalin (Tlc) was utilized as a protein scaffold to engineer an Anticalin that specifically binds and functionally blocks vascular endothelial growth factor A (VEGF-A), a pivotal inducer of physiological angiogenesis that also plays a crucial role in several neovascular diseases. Starting from a naive combinatorial library where residues that form the natural ligand-binding site of Tlc were randomized, followed by affinity maturation, the final Anticalin PRS-050 was selected to bind all major splice forms of VEGF-A with picomolar affinity. Moreover, this Anticalin cross-reacts with the murine ortholog. PRS-050 efficiently antagonizes the interaction between VEGF-A and its cellular receptors, and it inhibits VEGF-induced mitogenic signaling as well as proliferation of primary human endothelial cells with subnanomolar IC50 values. Intravitreal administration of the Anticalin suppressed VEGF-induced blood-retinal barrier breakdown in a rabbit model. To allow lasting systemic neutralization of VEGF-A in vivo, the plasma half-life of the Anticalin was extended by site-directed PEGylation. The modified Anticalin efficiently blocked VEGF-mediated vascular permeability as well as growth of tumor xenografts in nude mice, concomitantly with reduction in microvessel density. In contrast to bevacizumab, the Anticalin did not trigger platelet aggregation and thrombosis in human FcγRIIa transgenic mice, thus suggesting an improved safety profile. Since neutralization of VEGF-A activity is well known to exert beneficial effects in cancer and other neovascular diseases, including wet age-related macular degeneration, this Anticalin offers a novel potent small protein antagonist for differentiated therapeutic intervention in oncology and ophthalmology.


Assuntos
Lipocalinas/farmacologia , Terapia de Alvo Molecular , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Complexo Antígeno-Anticorpo/metabolismo , Bevacizumab/farmacologia , Bevacizumab/uso terapêutico , Barreira Hematorretiniana/patologia , Permeabilidade Capilar , Proliferação de Células/efeitos dos fármacos , Feminino , Meia-Vida , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lipocalinas/farmacocinética , Lipocalinas/uso terapêutico , Camundongos Nus , Camundongos Transgênicos , Mitógenos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Polietilenoglicóis/química , Engenharia de Proteínas , Coelhos , Receptores de IgG/metabolismo , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Trombose/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
PLoS One ; 10(11): e0142206, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26529243

RESUMO

Lipocalin-type prostaglandin D synthase (L-PGDS) is a member of the lipocalin superfamily, which is composed of secretory transporter proteins, and binds a wide variety of small hydrophobic molecules. Using this function, we have reported the feasibility of using L-PGDS as a novel drug delivery vehicle for poorly water-soluble drugs. In this study, we show the development of a drug delivery system using L-PGDS, one that enables the direct clinical use of 7-ethyl-10-hydroxy-camptothecin (SN-38), a poorly water-soluble anti-cancer drug. In the presence of 2 mM L-PGDS, the concentration of SN-38 in PBS increased 1,130-fold as compared with that in PBS. Calorimetric experiments revealed that L-PGDS bound SN-38 at a molecular ratio of 1:3 with a dissociation constant value of 60 µM. The results of an in vitro growth inhibition assay revealed that the SN-38/L-PGDS complexes showed high anti-tumor activity against 3 human cancer cell lines, i.e., Colo201, MDA-MB-231, and PC-3 with a potency similar to that of SN-38 used alone. The intravenous administration of SN-38/L-PGDS complexes to mice bearing Colo201 tumors showed a pronounced anti-tumor effect. Intestinal mucositis, which is one of the side effects of this drug, was not observed in mice administered SN-38/L-PGDS complexes. Taken together, L-PGDS enables the direct usage of SN-38 with reduced side effects.


Assuntos
Antineoplásicos , Camptotecina/análogos & derivados , Portadores de Fármacos , Oxirredutases Intramoleculares , Lipocalinas , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Camptotecina/farmacocinética , Camptotecina/farmacologia , Linhagem Celular Tumoral , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Oxirredutases Intramoleculares/farmacocinética , Oxirredutases Intramoleculares/farmacologia , Irinotecano , Lipocalinas/farmacocinética , Lipocalinas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Solubilidade
14.
Mol Cell Biochem ; 410(1-2): 207-13, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26350566

RESUMO

Over-activated PI3K/Akt signaling, a pathway strongly related to cancer survival and proliferation, has been reported recently to be involved in pulmonary artery smooth muscle cell apoptosis and proliferation in pulmonary hypertension (PH). In this study, we observed greatly increased lipocalin-2 (Lcn2) expression accompanied with over-activated PI3K/Akt signaling in a standard rat model of PH induced by monocrotaline. In view of the close relationship between Lcn2 and PI3K/Akt pathway, we hypothesized that the up-regulated Lcn2 might be a trigger of over-activated PI3K/Akt signaling in PH. Our results showed that Lcn2 significantly activated the PI3K/Akt pathway (determined by augmented Akt phosphorylation and up-regulated Mdm2) and significantly promoted proliferation (assessed by Ki67 staining) in cultured human pulmonary artery smooth muscle cells. Furthermore, we demonstrated that inhibition of Akt phosphorylation (LY294002) abrogated the Lcn2-promoted proliferation in cultured human pulmonary artery smooth muscle cells. In conclusion, Lcn2 significantly promoted human pulmonary artery smooth muscle cell proliferation by activating PI3K/Akt pathway. Further study on the role and mechanism of Lcn2 will help explore novel therapeutic strategies based on attenuating over-activated PI3K/Akt signaling in PH.


Assuntos
Proteínas de Fase Aguda/farmacologia , Proliferação de Células/efeitos dos fármacos , Lipocalinas/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Remodelação Vascular/efeitos dos fármacos , Animais , Células Cultivadas , Modelos Animais de Doenças , Ativação Enzimática , Humanos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/enzimologia , Hipertensão Pulmonar/patologia , Lipocalina-2 , Lipocalinas/metabolismo , Masculino , Monocrotalina , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Artéria Pulmonar/enzimologia , Artéria Pulmonar/patologia , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima
15.
Nat Commun ; 6: 8330, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26390966

RESUMO

Cyclic diguanylate monophosphate (c-di-GMP) is a well-conserved second messenger in bacteria. During infection, the innate immune system can also sense c-di-GMP; however, whether bacterial pathogens utilize c-di-GMP as a weapon to fight against host defense for survival and possible mechanisms underlying this process remain poorly understood. Siderocalin (LCN2) is a key antibacterial component of the innate immune system and sequesters bacterial siderophores to prevent acquisition of iron. Here we show that c-di-GMP can directly target the human LCN2 protein to inhibit its antibacterial activity. We demonstrate that c-di-GMP specifically binds to LCN2. In addition, c-di-GMP can compete with bacterial ferric siderophores to bind LCN2. Furthermore, c-di-GMP can significantly reduce LCN2-mediated inhibition on the in vitro growth of Escherichia coli. Thus, LCN2 acts as a c-di-GMP receptor. Our findings provide insight into the mechanism by which bacteria utilize c-di-GMP to interfere with the innate immune system for survival.


Assuntos
Proteínas de Fase Aguda/metabolismo , Antibacterianos/metabolismo , Proteínas de Transporte/metabolismo , Lipocalinas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/farmacologia , Proteínas de Transporte/genética , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Bases de Dados Factuais , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação da Expressão Gênica , Humanos , Lipocalina-2 , Lipocalinas/genética , Lipocalinas/farmacologia , Modelos Químicos , Modelos Moleculares , Estrutura Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Recombinantes
16.
Hypertension ; 66(1): 158-66, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25987661

RESUMO

Activation of the mineralocorticoid receptor has been shown to be deleterious in cardiovascular diseases (CVDs). We have recently shown that lipocalin 2 (Lcn2), or neutrophil gelatinase-associated lipocalin (NGAL), is a primary target of aldosterone/mineralocorticoid receptor in the cardiovascular system. Lcn2 is a circulating protein, which binds matrix metalloproteinase 9 and modulates its stability. We hypothesized that Lcn2 could be a mediator of aldosterone/mineralocorticoid receptor profibrotic effects in the cardiovascular system. Correlations between aldosterone and profibrotic markers, such as procollagen type I N-terminal peptide, were investigated in healthy subjects and subjects with abdominal obesity. The implication of Lcn2 in the mineralocorticoid pathway was studied using Lcn2 knockout mice subjected to a nephrectomy/aldosterone/salt (NAS) challenge for 4 weeks. In human subjects, NGAL/matrix metalloproteinase 9 was positively correlated with plasma aldosterone and fibrosis biomarkers. In mice, loss of Lcn2 prevented the NAS-induced increase of plasma procollagen type I N-terminal peptide, as well as the increase of collagen fibers deposition and collagen I expression in the coronary vessels and the aorta. The lack of Lcn2 also blunted the NAS-induced increase in systolic blood pressure. Ex vivo, treatment of human fibroblasts with recombinant Lcn2 induced the expression of collagen I and the profibrotic galectin-3 and cardiotrophin-1 molecules. Our results showed that Lcn2 plays a key role in aldosterone/mineralocorticoid receptor-mediated vascular fibrosis. The clinical data indicate that this may translate in human patients. Lcn2 is, therefore, a new biotarget in cardiovascular fibrosis induced by mineralocorticoid activation.


Assuntos
Proteínas de Fase Aguda/fisiologia , Aldosterona/toxicidade , Lipocalinas/fisiologia , Obesidade Abdominal/fisiopatologia , Proteínas Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas de Fase Aguda/deficiência , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/farmacologia , Aldosterona/sangue , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Cardiomiopatia Hipertrófica/induzido quimicamente , Cardiomiopatia Hipertrófica/fisiopatologia , Células Cultivadas , Citocinas/biossíntese , Citocinas/genética , Feminino , Fibroblastos , Fibrose , Galectina 3/biossíntese , Galectina 3/sangue , Galectina 3/genética , Humanos , Hipertensão/fisiopatologia , Hipertrofia , Rim/patologia , Lipocalina-2 , Lipocalinas/sangue , Lipocalinas/genética , Lipocalinas/farmacologia , Masculino , Camundongos , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Nefrectomia/efeitos adversos , Obesidade Abdominal/sangue , Proteínas Oncogênicas/deficiência , Proteínas Oncogênicas/genética , Fragmentos de Peptídeos/sangue , Pró-Colágeno/sangue , Proteínas Proto-Oncogênicas/sangue , Proteínas Proto-Oncogênicas/farmacologia , Ratos , Proteínas Recombinantes/farmacologia
17.
Nephrology (Carlton) ; 20(9): 646-53, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25943501

RESUMO

AIM: The aim of this study was to investigate the effect of neutrophil gelatinase-associated lipocalin (NGAL) on the rat cecal ligation and puncture (CLP)-induced sepsis and the possible mechanism. METHODS: Thirty male Sprague-Dawley rats underwent CLP as sepsis models and were randomized into three groups including the sham-operated group (sham, n = 10), which only underwent a laparotomy; the sepsis group (sepsis, n = 10), which underwent CLP and subcutaneous injection of normal saline; and the sepsis + NGAL group (sepsis + NGAL, n = 10), which underwent CLP and subcutaneous injection of NGAL. Urine, blood and kidney tissue samples were collected for the determination of urine NGAL (uNGAL), plasma NGAL (pNGAL), serum creatinine (Scr), blood urea nitrogen (BUN), histomorphological and immunohistochemical examination, lipid peroxidation product malondialdehyde (MDA) and superoxide dismutase (SOD), and expression of heme oxygenase-1 (HO)-1. RESULTS: The levels of uNGAL, pNGAL, Scr, BUN, kidney injury score, positive TUNEL staining, activated Caspase-3 and Bax, and kidney tissue MDA levels in the sepsis group were significantly increased compared with those in the sham-operated group and the sepsis + NGAL group (P < 0.05). SOD level and HO-1 expression in sepsis + NGAL group were significantly higher than those in the sham-operated group and the sepsis group (P < 0.05). CONCLUSION: NGAL can attenuate kidney injury and apoptosis in the rat CLP model of sepsis. And the protective effect of NGAL was probably due to the inhibition of apoptosis and lipid peroxidation, and increased expression of HO-1.


Assuntos
Injúria Renal Aguda/prevenção & controle , Apoptose/efeitos dos fármacos , Ceco/cirurgia , Rim/efeitos dos fármacos , Lipocalinas/farmacologia , Sepse/tratamento farmacológico , Injúria Renal Aguda/microbiologia , Injúria Renal Aguda/patologia , Animais , Ceco/microbiologia , Citoproteção , Modelos Animais de Doenças , Heme Oxigenase (Desciclizante)/metabolismo , Injeções Subcutâneas , Rim/patologia , Ligadura , Peroxidação de Lipídeos/efeitos dos fármacos , Lipocalinas/administração & dosagem , Masculino , Malondialdeído/metabolismo , Punções , Ratos Sprague-Dawley , Sepse/microbiologia , Sepse/patologia , Superóxido Dismutase/metabolismo , Fatores de Tempo
18.
J Allergy Clin Immunol ; 136(4): 1047-54.e10, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25843313

RESUMO

BACKGROUND: The recently identified dog lipocalin allergen Can f 4 is an important respiratory allergen. OBJECTIVE: We sought to comprehensively characterize the memory CD4(+) T-cell responses of allergic and nonallergic subjects to Can f 4. METHODS: Can f 4-specific CD4(+)CD45RO(+) T-cell lines (TCLs) from allergic and healthy subjects were established and characterized by their functional and phenotypic properties. The epitope specificity of the TCLs was tested with 48 overlapping 16-mer peptides spanning the sequence of Can f 4. HLA restriction of the specific TCLs and the binding capacity of the epitope-containing peptides to common HLA class II molecules were studied. RESULTS: Can f 4-specific memory CD4(+) TCLs were obtained at an 8-fold higher frequency from allergic than from nonallergic subjects. Functionally, the TCLs of allergic subjects exhibited a higher T-cell receptor avidity and expression of CD25 and predominantly produced IL-4 and IL-5. The TCLs of nonallergic subjects mostly secreted IFN-γ and IL-10, with high CXCR3 expression. Several distinct T-cell epitope regions along the allergen were identified. Importantly, the peptides from the region between amino acids 43 and 67 showed promiscuous HLA-binding capacity and induced memory CD4(+) T-cell responses in 90% of the allergic donors. CONCLUSION: Productive TH2-deviated memory T-cell responses to Can f 4 are observed in allergic but not nonallergic subjects. A 19-mer peptide sequence covering the core of the immunodominant region of the allergen is a potential target for the development of peptide-based allergen immunotherapy.


Assuntos
Alérgenos/imunologia , Hipersensibilidade/imunologia , Memória Imunológica , Lipocalinas/imunologia , Células Th2/imunologia , Alérgenos/farmacologia , Animais , Linhagem Celular , Citocinas/imunologia , Cães , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Hipersensibilidade/patologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Lipocalinas/farmacologia , Masculino , Receptores CXCR3/imunologia
19.
J Neurochem ; 132(6): 622-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25557118

RESUMO

Inflammation is a key part of central nervous system pathophysiology. However, inflammatory factors are now thought to have both beneficial and deleterious effects. Here, we examine the hypothesis that lipocalin-2 (LCN2), an inflammatory molecule that can be up-regulated in the distressed central nervous system, may enhance angiogenesis in brain endothelial cells. Adding LCN2 (0.5-2.0 µg/mL) to RBE (Rat brain endothelial cells). 4 rat brain endothelial cells significantly increased matrigel tube formation and scratch migration, and also elevated levels of iron and reactive oxygen species. Co-treatment with a radical scavenger (U83836E), a Nox inhibitor (apocynin) and an iron chelating agent (deferiprone) significantly dampened the ability of LCN2 to enhance tube formation and scratch migration in brain endothelial cells. These findings provide in vitro proof of the concept that LCN2 can promote angiogenesis via iron- and reactive oxygen species-related pathways, and support the idea that LCN2 may contribute to the neurovascular recovery aspects of inflammation. Angiogenesis is an important part of stroke recovery. In the present study, we examined the hypothesis that lipocalin-2 (LCN2) may enhance angiogenesis in brain endothelial cells. LCN2 promoted tube formation and migration via iron and ROS-related pathways in rat brain endothelial cells. ROS scavengers, Nox inhibitors and iron chelators all dampened the ability of LCN2 to enhance in vitro angiogenesis. These findings support the idea that LCN2 that is released by damaged neurons may act as a 'help me' signal that promotes neurovascular recovery after stroke and brain injury.


Assuntos
Encéfalo/metabolismo , Células Endoteliais/metabolismo , Ferro/metabolismo , Lipocalinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Lipocalina-2 , Ratos
20.
Int J Cancer ; 137(5): 1235-44, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25516376

RESUMO

The antitumor activity of prostaglandin (PG) D2 has been demonstrated against some types of cancer, including gastric cancer. However, exogenous PGD2 is not useful from a clinical point of view because it is rapidly metabolized in vivo. The aim of this study was to clarify the antitumor efficacy of an alternative, PGD synthase (PGDS), on gastric cancer cells. The effects of PGD2 and PGDS on the proliferation of gastric cancer cells were examined in vivo and in vitro. The expression levels of PGD2 receptors and peroxisome proliferator-activated receptor γ (PPARγ) were evaluated by RT-PCR. The effects of a PPARγ antagonist or siPPARγ on the proliferation of cancer cells and the c-myc and cyclin D1 expression were examined in the presence or absence of PGD2 or PGDS. PPARγ was expressed in gastric cancer cell lines, but PGD2 receptors were not. PGD2 and PGDS significantly decreased the proliferation of gastric cancer cells that highly expressed PPARγ. PGDS increased the PGD2 production of gastric cancer cells. A PPARγ antagonist and siPPARγ transfection significantly suppressed the growth-inhibitory effects of PGD2 and PGDS. Expression of c-myc and cyclin D1 was significantly decreased by PGD2 ; this inhibitory effect was suppressed by PPARγ antagonist. Both PGD2 and PGDS significantly decreased subcutaneous tumor growth in vivo. Tumor volume after PGDS treatment was significantly less than PGD2 treatment. These findings suggest that PGDS and PGD2 decrease the proliferation of gastric cancer cells through PPARγ signaling. PGDS is a potentially promising therapeutic agent for gastric cancers that express PPARγ.


Assuntos
Antineoplásicos/administração & dosagem , Oxirredutases Intramoleculares/administração & dosagem , Lipocalinas/administração & dosagem , PPAR gama/genética , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Oxirredutases Intramoleculares/farmacologia , Lipocalinas/farmacologia , Camundongos , PPAR gama/metabolismo , Prostaglandina D2/administração & dosagem , Prostaglandina D2/farmacologia , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...