Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Immune Netw ; 24(2): e17, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38725672

RESUMEN

We have reported that anterior cruciate ligament (ACL) injury leads to the differential dysregulation of the complement system in the synovium as compared to meniscus tear (MT) and proposed this as a mechanism for a greater post-injury prevalence of post traumatic osteoarthritis (PTOA). To explore additional roles of complement proteins and regulators, we determined the presence of decay-accelerating factor (DAF), C5b, and membrane attack complexes (MACs, C5b-9) in discarded surgical synovial tissue (DSST) collected during arthroscopic ACL reconstructive surgery, MT-related meniscectomy, osteoarthritis (OA)-related knee replacement surgery and normal controls. Multiplexed immunohistochemistry was used to detect and quantify complement proteins. To explore the involvement of body mass index (BMI), after these 2 injuries, we examined correlations among DAF, C5b, MAC and BMI. Using these approaches, we found that synovial cells after ACL injury expressed a significantly lower level of DAF as compared to MT (p<0.049). In contrast, C5b staining synovial cells were significantly higher after ACL injury (p<0.0009) and in OA DSST (p<0.039) compared to MT. Interestingly, there were significantly positive correlations between DAF & C5b (r=0.75, p<0.018) and DAF & C5b (r=0.64 p<0.022) after ACL injury and MT, respectively. The data support that DAF, which should normally dampen C5b deposition due to its regulatory activities on C3/C5 convertases, does not appear to exhibit that function in inflamed synovia following either ACL injury or MT. Ineffective DAF regulation may be an additional mechanism by which relatively uncontrolled complement activation damages tissue in these injury states.

2.
Nat Nanotechnol ; 19(2): 246-254, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37798566

RESUMEN

Effective inhibition of the complement system is needed to prevent the accelerated clearance of nanomaterials by complement cascade and inflammatory responses. Here we show that a fusion construct consisting of human complement receptor 2 (CR2) (which recognizes nanosurface-deposited complement 3 (C3)) and complement receptor 1 (CR1) (which blocks C3 convertases) inhibits complement activation with picomolar to low nanomolar efficacy on many types of nanomaterial. We demonstrate that only a small percentage of nanoparticles are randomly opsonized with C3 both in vitro and in vivo, and CR2-CR1 immediately homes in on this subpopulation. Despite rapid in vivo clearance, the co-injection of CR2-CR1 in rats, or its mouse orthologue CR2-Crry in mice, with superparamagnetic iron oxide nanoparticles nearly completely blocks complement opsonization and unwanted granulocyte/monocyte uptake. Furthermore, the inhibitor completely prevents lethargy caused by bolus-injected nanoparticles, without inducing long-lasting complement suppression. These findings suggest the potential of the targeted complement regulators for clinical evaluation.


Asunto(s)
Nanopartículas , Receptores de Complemento 3d , Ratas , Ratones , Humanos , Animales , Receptores de Complemento 3b , Activación de Complemento , Complemento C3 , Proteínas Recombinantes de Fusión
4.
ACS Nano ; 2022 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-35507641

RESUMEN

Many aspects of innate immune responses to SARS viruses remain unclear. Of particular interest is the role of emerging neutralizing antibodies against the receptor-binding domain (RBD) of SARS-CoV-2 in complement activation and opsonization. To overcome challenges with purified virions, here we introduce "pseudovirus-like" nanoparticles with ∼70 copies of functional recombinant RBD to map complement responses. Nanoparticles fix complement in an RBD-dependent manner in sera of all vaccinated, convalescent, and naïve donors, but vaccinated and convalescent donors with the highest levels of anti-RBD antibodies show significantly higher IgG binding and higher deposition of the third complement protein (C3). The opsonization via anti-RBD antibodies is not an efficient process: on average, each bound antibody promotes binding of less than one C3 molecule. C3 deposition is exclusively through the alternative pathway. C3 molecules bind to protein deposits, but not IgG, on the nanoparticle surface. Lastly, "pseudovirus-like" nanoparticles promote complement-dependent uptake by granulocytes and monocytes in the blood of vaccinated donors with high anti-RBD titers. Using nanoparticles displaying SARS-CoV-2 proteins, we demonstrate subject-dependent differences in complement opsonization and immune recognition.

5.
Front Immunol ; 11: 575154, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178202

RESUMEN

Natural IgM antibodies (NAbs) have been shown to recognize injury-associated neoepitopes and to initiate pathogenic complement activation. The NAb termed C2 binds to a subset of phospholipids displayed on injured cells, and its role(s) in arthritis, as well as the potential therapeutic benefit of a C2 NAb-derived ScFv-containing protein fused to a complement inhibitor, complement receptor-related y (Crry), on joint inflammation are unknown. Our first objective was to functionally test mAb C2 binding to apoptotic cells from the joint and also evaluate its inflammation enhancing capacity in collagen antibody-induced arthritis (CAIA). The second objective was to generate and test the complement inhibitory capacity of C2-Crry fusion protein in the collagen-induced arthritis (CIA) model. The third objective was to demonstrate in vivo targeting of C2-Crry to damaged joints in mice with arthritis. The effect of C2-NAb on CAIA in C57BL/6 mice was examined by inducing a suboptimal disease. The inhibitory effect of C2-Crry in DBA/1J mice with CIA was determined by injecting 2x per week with a single dose of 0.250 mg/mouse. Clinical disease activity (CDA) was examined, and knee joints were fixed for analysis of histopathology, C3 deposition, and macrophage infiltration. In mice with suboptimal CAIA, at day 10 there was a significant (p < 0.017) 74% increase in the CDA in mice treated with C2 NAb, compared to mice treated with F632 control NAb. In mice with CIA, at day 35 there was a significant 39% (p < 0.042) decrease in the CDA in mice treated with C2-Crry. Total scores for histopathology were also 50% decreased (p < 0.0005) in CIA mice treated with C2-Crry. C3 deposition was significantly decreased in the synovium (44%; p < 0.026) and on the surface of cartilage (42%; p < 0.008) in mice treated with C2-Crry compared with PBS treated CIA mice. Furthermore, C2-Crry specifically bound to apoptotic fibroblast-like synoviocytes in vitro, and also localized in the knee joints of arthritic mice as analyzed by in vivo imaging. In summary, NAb C2 enhanced arthritis-related injury, and targeted delivery of C2-Crry to inflamed joints demonstrated disease modifying activity in a mouse model of human inflammatory arthritis.


Asunto(s)
Antirreumáticos/farmacología , Artritis Experimental/tratamiento farmacológico , Activación de Complemento/efectos de los fármacos , Inmunoglobulina M/farmacología , Articulaciones/efectos de los fármacos , Receptores de Complemento 3b/metabolismo , Anticuerpos de Cadena Única/farmacología , Sinoviocitos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Experimental/patología , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Articulaciones/inmunología , Articulaciones/metabolismo , Articulaciones/patología , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Proteínas Recombinantes de Fusión/farmacología , Sinoviocitos/inmunología , Sinoviocitos/metabolismo , Sinoviocitos/patología , Timocitos/efectos de los fármacos , Timocitos/inmunología , Timocitos/metabolismo , Timocitos/patología
6.
Front Immunol ; 11: 201, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32153567

RESUMEN

The complement system plays an important role in the pathogenesis of rheumatoid arthritis (RA). Besides driving lectin pathway (LP) activation, the mannan-binding lectin (MBL)-associated serine proteases (MASPs) also play a key role in regulating the alternative pathway (AP). We evaluated the effects of N-acetylgalactosamine (GalNAc)-conjugated MASP-1 and MASP-2 duplexes in vitro and in mice with and without arthritis to examine whether knockdown of MASP-1 and MASP-2 expression affects the development of arthritis. GalNAc-siRNAs for MASP-1 and MASP-2 demonstrated robust silencing of MASP-1 or MASP-2 at pM concentrations in vitro. To evaluate the impact of silencing in arthritic mice, we used the collagen antibody-induced arthritis (CAIA) mouse model of RA. Mice were injected a 10 mg/kg dose of GalNAc-siRNAs 3x s.q. prior to the induction of CAIA. Liver gene expression was examined using qRT-PCR, and protein levels were confirmed in the circulation by sandwich immunoassays and Western blot. At day 10, CAIA mice separately treated with MASP-1 and MASP-2 duplexes had a specific reduction in expression of liver MASP-1 (70-95%, p < 0.05) and MASP-2 (90%, p < 0.05) mRNA, respectively. MASP-1-siRNA treatment resulted in a 95% reduction in levels of MASP-1 protein in circulation with no effect on MASP-2 levels and clinical disease activity (CDA). In mice injected with MASP-2 duplex, there was a significant (p < 0.05) 90% decrease in ex vivo C4b deposition on mannan, with nearly complete elimination of MASP-2 in the circulation. MASP-2 silencing initially significantly decreased CDA by 60% but subsequently changed to a 40% decrease vs. control. Unexpectedly, GalNAc-siRNA-mediated knockdown of MASP-1 and MASP-2 revealed a marked effect of these proteins on the transcription of FD under normal physiological conditions, whereas LPS-induced inflammatory conditions reversed this effect on FD levels. LPS is recognized by Toll-like receptor 4 (TLR4), we found MBL not only binds to TLR4 an interaction with a Kd of 907 nM but also upregulated FD expression in differentiated adipocytes. We show that MASP-2 knockdown impairs the development of RA and that the interrelationship between proteins of the LP and the AP may extend to the transcriptional modulation of the FD gene.


Asunto(s)
Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Factor D del Complemento/metabolismo , Vía Alternativa del Complemento/genética , Lectina de Unión a Manosa de la Vía del Complemento/genética , Transcripción Genética/genética , Animales , Factor D del Complemento/genética , Expresión Génica , Lipopolisacáridos/farmacología , Hígado/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección
7.
Mol Pharm ; 16(10): 4274-4281, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31556296

RESUMEN

Feraheme (ferumoxytol), a negatively charged, carboxymethyl dextran-coated ultrasmall superparamagnetic iron oxide nanoparticle (USPIO, 30 nm, -16 mV), is clinically approved as an iron supplement and is used off-label for magnetic resonance imaging (MRI) of macrophage-rich lesions, but the mechanism of recognition is not known. We investigated mechanisms of uptake of Feraheme by various types of macrophages in vitro and in vivo. The uptake by mouse peritoneal macrophages was not inhibited in complement-deficient serum. In contrast, the uptake of larger and less charged SPIO nanoworms (60 nm, -5 mV; 120 nm, -5 mV, respectively) was completely inhibited in complement deficient serum, which could be attributed to more C3 molecules bound per nanoparticle than Feraheme. The uptake of Feraheme in vitro was blocked by scavenger receptor (SR) inhibitor polyinosinic acid (PIA) and by antibody against scavenger receptor type A I/II (SR-AI/II). Antibodies against other SRs including MARCO, CD14, SR-BI, and CD11b had no effect on Feraheme uptake. Intraperitoneally administered PIA inhibited the peritoneal macrophage uptake of Feraheme in vivo. Nonmacrophage cells transfected with SR-AI plasmid efficiently internalized Feraheme but not noncharged ultrasmall SPIO of the same size (26 nm, -6 mV), suggesting that the anionic carboxymethyl groups of Feraheme are responsible for the SR-AI recognition. The uptake by nondifferentiated bone marrow derived macrophages (BMDM) and by BMDM differentiated into M1 (proinflammatory) and M2 (anti-inflammatory) types was efficiently inhibited by PIA and anti-SR-AI/II antibody. Interestingly, all BMDM types expressed similar levels of SR-AI/II. In conclusion, Feraheme is efficiently recognized via SR-AI/II but not via complement by different macrophage types. The recognition by the common phagocytic receptor has implications for specificity of imaging of macrophage subtypes.


Asunto(s)
Óxido Ferrosoférrico/metabolismo , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Receptores Depuradores de Clase A/metabolismo , Animales , Células Cultivadas , Femenino , Hematínicos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C
8.
Mol Pharm ; 16(6): 2445-2451, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31091104

RESUMEN

Diseases of the joints affect over 10% of the world's population, resulting in significant morbidity. There is an unmet need in strategies for specific delivery of therapeutics to the joints. Collagen type II is synthesized by chondrocytes and is mainly restricted to the cartilage and tendons. Arthrogen-CIA is a commercially available anticollagen II antibody cocktail that reacts with 5 different epitopes on human, bovine, and mouse collagen II. Arthrogen has been used for induction of experimental rheumatoid arthritis (RA) in mice because of high complement activation on the cartilage surface. Native collagen II might serve as a useful target for potential delivery of therapeutics to the joint. To evaluate the efficiency and specificity of targeting collagen II, Arthrogen was labeled with near-infrared (NIR) dye IRDye 800 or IRDye 680. Using ex vivo NIR imaging, we demonstrate that Arthrogen efficiently and specifically accumulated in the limb joints regardless of the label dye or injection route (intravenous and subcutaneous). After subcutaneous injection, the mean fluorescence of the hind limb joints was 19 times higher than that of the heart, 8.7 times higher than that of the liver, and 3.7 times higher than that of the kidney. Control mouse IgG did not show appreciable accumulation. Microscopically, the antibody accumulated on the cartilage surface of joints and on endosteal surfaces. A monoclonal antibody against a single epitope of collagen II showed similar binding affinity and elimination half-life, but about three times lower targeting efficiency than Arthrogen in vitro and ex vivo, and about two times lower targeting efficiency in vivo. We suggest that an antibody against multiple epitopes of collagen II could be developed into a highly effective and specific targeting strategy for diseases of the joints or spine.


Asunto(s)
Anticuerpos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Colágeno Tipo II/inmunología , Animales , Anticuerpos/inmunología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Cartílago/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos BALB C
9.
Immunohorizons ; 2(8): 274-295, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30417171

RESUMEN

Mannan-binding lectin-associated serine protease 3 (MASP-3) regulates the alternative pathway of complement and is predominantly synthesized in the liver. The role of liver-derived MASP-3 in the pathogenesis of rheumatoid arthritis (RA) is unknown. We hypothesized that liver-derived MASP-3 is essential for the development of joint damage and that targeted inhibition of MASP-3 in the liver can attenuate arthritis. We used MASP-3-specific small interfering RNAs (siRNAs) conjugated to N-acetylgalactosamine (GalNAc) to specifically target the liver via asialoglycoprotein receptors. Active GalNAc-MASP3-siRNA conjugates were identified, and in vivo silencing of liver MASP-3 mRNA was demonstrated in healthy mice. The s.c. treatment with GalNAc-MASP-3-siRNAs specifically decreased the expression of MASP-3 in the liver and the level of MASP-3 protein in circulation of mice without affecting the levels of the other spliced products. In mice with collagen Ab-induced arthritis, s.c. administration of GalNAc-MASP-3-siRNA decreased the clinical disease activity score to 50% of controls, with decrease in histopathology scores and MASP-3 deposition. To confirm the ability to perform MASP-3 gene silencing in human cells, we generated a lentivirus expressing a short hairpin RNA specific for human MASP-3 mRNA. This procedure not only eliminated the short-term (at day 15) expression of MASP-3 in HepG2 and T98G cell lines but also diminished the long-term (at day 60) synthesis of MASP-3 protein in T98G cells. Our study demonstrates that isoform-specific silencing of MASP-3 in vivo modifies disease activity in a mouse model of RA and suggests that liver-directed MASP3 silencing may be a therapeutic approach in human RA.

10.
J Immunol ; 199(5): 1835-1845, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28739878

RESUMEN

Complement plays an important role in the pathogenesis of rheumatoid arthritis. Although the alternative pathway (AP) is known to play a key pathogenic role in models of rheumatoid arthritis, the importance of the lectin pathway (LP) pattern recognition molecules such as ficolin (FCN) A, FCN B, and collectin (CL)-11, as well as the activating enzyme mannose-binding lectin-associated serine protease-2 (MASP-2), are less well understood. We show in this article that FCN A-/- and CL-11-/- mice are fully susceptible to collagen Ab-induced arthritis (CAIA). In contrast, FCN B-/- and MASP-2-/-/sMAp-/- mice are substantially protected, with clinical disease activity decreased significantly (p < 0.05) by 47 and 70%, respectively. Histopathology scores, C3, factor D, FCN B deposition, and infiltration of synovial macrophages and neutrophils were similarly decreased in FCN B-/- and MASP-2-/-/sMAp-/- mice. Our data support that FCN B plays an important role in the development of CAIA, likely through ligand recognition in the joint and MASP activation, and that MASP-2 also contributes to the development of CAIA, likely in a C4-independent manner. Decreased AP activity in the sera from FCN B-/- and MASP-2-/-/sMAp-/- mice with arthritis on adherent anti-collagen Abs also support the hypothesis that pathogenic Abs, as well as additional inflammation-related ligands, are recognized by the LP and operate in vivo to activate complement. Finally, we also speculate that the residual disease seen in our studies is driven by the AP and/or the C2/C4 bypass pathway via the direct cleavage of C3 through an LP-dependent mechanism.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Lectina de Unión a Manosa de la Vía del Complemento , Inflamación/inmunología , Lectinas/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Células Cultivadas , Colágeno/inmunología , Colectinas/genética , Colectinas/metabolismo , Proteínas del Sistema Complemento/metabolismo , Humanos , Lectinas/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ficolinas
11.
J Immunol ; 197(9): 3680-3694, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27707997

RESUMEN

The complement system is proposed to play an important role in the pathogenesis of rheumatoid arthritis (RA). The complement system mannan-binding lectin-associated serine proteases (MASP)-1/3 cleave pro-factor D (proDf; inactive) into Df (active), but it is unknown where this cleavage occurs and whether inhibition of MASP-1/3 is a relevant therapeutic strategy for RA. In the present study, we show that the cleavage of proDf into Df by MASP-1/3 can occur in the circulation and that inhibition of MASP-1/3 by gene silencing is sufficient to ameliorate collagen Ab-induced arthritis in mice. Specifically, to examine the cleavage of proDf into Df, MASP-1/3-producing Df-/- liver tissue (donor) was transplanted under the kidney capsule of MASP-1/3-/- (recipient) mice. Five weeks after the liver transplantation, cleaved Df was present in the circulation of MASP-1/3-/- mice. To determine the individual effects of MASP-1/3 and Df gene silencing on collagen Ab-induced arthritis, mice were injected with scrambled, MASP-1/3-targeted, or Df-targeted small interfering RNAs (siRNAs). The mRNA levels for MASP-1 and -3 decreased in the liver to 62 and 58%, respectively, in mice injected with MASP-1/3 siRNAs, and Df mRNA decreased to 53% in the adipose tissue of mice injected with Df siRNAs; additionally, circulating MASP-1/3 and Df protein levels were decreased. In mice injected with both siRNAs the clinical disease activity, histopathologic injury scores, C3 deposition, and synovial macrophage/neutrophil infiltration were significantly decreased. Thus, MASP-1/3 represent a new therapeutic target for the treatment of RA, likely through both direct effects on the lectin pathway and indirectly through the alternative pathway.


Asunto(s)
Artritis Experimental/terapia , Artritis Reumatoide/terapia , Factor D del Complemento/metabolismo , Lectina de Unión a Manosa de la Vía del Complemento , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Interferencia de ARN , Animales , Artritis Experimental/genética , Artritis Reumatoide/genética , Factor D del Complemento/genética , Humanos , Masculino , Lectina de Unión a Manosa/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteolisis , ARN Interferente Pequeño/genética
12.
J Immunol ; 194(11): 5446-54, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917104

RESUMEN

Rheumatoid arthritis (RA) is an inflammatory autoimmune joint disease in which the complement system plays an important role. Of the several components of complement, current evidence points to C5 as the most important inducer of inflammation. Several groups generated Abs or small interfering RNAs (siRNAs) or small molecule inhibitors against C5 and C5aR1 (CD88) that have showed some efficacy in RA in animal models. However, none of these candidate therapeutics has moved from bench to bedside. In this study, we test in collagen Ab-induced arthritis (CAIA) a new therapeutic strategy using a novel anti-C5ab-C5 siRNA conjugate. We first demonstrate that although C5aR2 or C5L2 (GPR77) plays no role in CAIA, C5aR1 contributes to pathogenesis. We demonstrate that injection of siRNAs blocking C5, C5aR1, or the combination decreased clinical disease activity in mice with CAIA by 45%, 51%, and 58%, respectively. Anti-C5 Ab (BB5.1) has only limited efficacy, but significantly reduced arthritis up to 66%. We then generated a novel anti-C5aR1 Ab-protamine-C5 siRNA conjugate. To our knowledge, we show for the first time that whereas unconjugated Ab plus siRNAs reduce arthritis by 19%, our anti-C5aR1 Ab-protamine-C5 siRNA conjugate was effective in reducing arthritis by 83% along with a parallel decrease in histopathology, C3 deposition, neutrophils, and macrophages in the joints of mice with CAIA. These data suggest that by targeting anti-C5 siRNAs to the receptor for its C5a activation fragment (C5aR1), a striking clinical effect can be realized.


Asunto(s)
Anticuerpos Antiidiotipos/uso terapéutico , Artritis Experimental/terapia , Artritis Reumatoide/terapia , Complemento C5/inmunología , ARN Interferente Pequeño/uso terapéutico , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Animales , Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Complemento C5/genética , Articulación de la Rodilla/citología , Articulación de la Rodilla/inmunología , Articulación de la Rodilla/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , ARN Mensajero/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Receptor de Anafilatoxina C5a/inmunología
13.
Mol Pharm ; 9(11): 3228-35, 2012 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-23003570

RESUMEN

Retinal pigment epithelium, which forms the outer blood-retinal barrier, is a critical barrier for transport of drugs to the retina. The purpose of this study was to develop a pigmented MDCK (P-MDCK) cell line as a rapidly established in vitro model for the outer blood-retinal barrier to assess the influence of melanin pigment on solute permeability. A melanin synthesizing P-MDCK cell line was developed by lentiviral transduction of human tyrosinase and p-protein genes in MDCK (NBL-2) cells. Melanin content, tyrosinase activity (conversion of L-dopa to dopachrome), and transepithelial electrical resistance (TEER) were measured. Expression of tyrosinase protein and p-protein in P-MDCK cells was confirmed by confocal microscopy. Effect of l-tyrosine (0 to 2 mM) in culture medium on melanin synthesis in P-MDCK cells was evaluated. Cell uptake and transepithelial transport of pigment-binding chloroquine (Log D = 1.59) and a negative control salicylic acid (Log D = -1.14) were investigated. P-MDCK cells expressed tyrosinase and p-protein. Tyrosinase activity was 4.5-fold higher in P-MDCK cells compared to wild type MDCK cells. The transepithelial electrical resistance stabilized by day 4 in both cell types, with the TEER being 958 ± 33 and 964 ± 58 Ω·cm(2) for P-MDCK and wild type cells, respectively. Melanin content in P-MDCK cells depended on the concentration of l-tyrosine in culture medium, and increased from 3 to 54 µg/mg protein with an increase in l-tyrosine content from 0 to 2 mM. When the cells were grown in 2 mM l-tyrosine, uptake of chloroquine was 2.3-fold higher and the transepithelial transport was 2.2-fold lower in P-MDCK cells when compared to wild type MDCK cells. No significant difference was observed for both cell uptake and transport of salicylic acid. We developed a P-MDCK cell line with tunable melanin synthesis as a rapidly developing surrogate for retinal pigment epithelium.


Asunto(s)
Barrera Hematorretinal/metabolismo , Cloroquina/metabolismo , Impedancia Eléctrica , Melaninas/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Ácido Salicílico/metabolismo , Tirosina/metabolismo , Animales , Transporte Biológico , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Células Cultivadas , Cromatografía Liquida , Perros , Humanos , Técnicas para Inmunoenzimas , Riñón/citología , Riñón/metabolismo , Levodopa/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Monofenol Monooxigenasa/metabolismo , Pigmentación , Espectrometría de Masas en Tándem
15.
Future Med Chem ; 4(6): 735-50, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22530638

RESUMEN

The immune response is replete with feedback control at many levels. These protective circuits are even functional within the arthritic joint, tempering disease to varying extents. An optimal therapy would inhibit autoimmune processes while maintaining protective circuitry. However, many of the cells and proteins that serve as important mediators of disease progression also play an active role in these protective circuits. The hypothesis considered in this review is that the inadvertent inhibition of protective circuitry adversely affects efficacy. Conversely, if therapeutics can be designed, which avoid inhibiting known regulatory circuits, efficacy will be improved. Understanding where these processes share signaling molecules will be crucial to the development of the next generation of therapeutics. This review discusses three well-defined signal transduction cascades; IL-2, IFNγ and TNF-α, and demonstrate within two cell types, T cells and macrophages, how these cytokines may contribute both to protection and to disease progression.


Asunto(s)
Artritis Reumatoide/inmunología , Interferón gamma/inmunología , Interleucina-2/inmunología , Macrófagos/inmunología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Artritis Reumatoide/patología , Progresión de la Enfermedad , Humanos , Macrófagos/patología , Transducción de Señal , Linfocitos T/patología
16.
Nanomedicine (Lond) ; 6(10): 1669-82, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22087799

RESUMEN

AIM: To develop and characterize an RGD peptide functionalized poly(lactide-co-glycolytic) acid (PLGA) nanosystem to deliver a STAT1 siRNA to joint tissues in a mouse model of rheumatoid arthritis. METHODS: RGD-PLGA polymer was synthesized and used in preparing functionalized nanoparticles loaded with either tracking material or siRNA. The properties of the nanoparticles and stability of siRNA after encapsulation was assessed. Nanoparticle distribution was determined both noninvasively and based on analysis of dissected organs from arthritic and healthy mice. Arthritic mice were treated with weekly doses of STAT1 siRNA-loaded nanoparticles or controls. Clinical disease was assessed. Paws of arthritic mice were sectioned for histology or processed for RNA. STAT1, Mrc-1, and IL-10 mRNA abundance was determined by quantitative PCR. RESULTS: Nanoparticles protected the siRNA from serum degradation. The presence of RGD peptide on the nanoparticles increased paw tissue uptake in arthritic mice. Furthermore, RGD functionalization increased lung delivery of nanoparticles in arthritic mice but not in control mice. Disease regressed in the STAT1 siRNA-treated animals and progressed in all control groups. STAT1 mRNA levels were decreased in paws of treated animals, while Mrc-1 and IL-10 mRNA levels were increased. CONCLUSION: RGD functionalized PLGA nanoparticles encapsulating STAT1-targeted siRNAs are efficacious in the treatment of established arthritis, possibly through a selective inhibition of macrophage and dendritic cell activation.


Asunto(s)
Artritis Reumatoide/terapia , Portadores de Fármacos/química , Portadores de Fármacos/síntesis química , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Factor de Transcripción STAT1/administración & dosificación , Animales , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Estabilidad de Medicamentos , Silenciador del Gen , Terapia Genética , Interleucina-10/metabolismo , Ácido Láctico/síntesis química , Ácido Láctico/química , Macrófagos/metabolismo , Ratones , Nanopartículas/química , Nanopartículas/ultraestructura , Oligopéptidos/síntesis química , Oligopéptidos/química , Ácido Poliglicólico/síntesis química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo
17.
PLoS One ; 6(9): e24616, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21915354

RESUMEN

BACKGROUND: P23H rhodopsin, a mutant rhodopsin, is known to aggregate and cause retinal degeneration. However, its effects on retinal pigment epithelial (RPE) cells are unknown. The purpose of this study was to determine the effect of P23H rhodopsin in RPE cells and further assess whether LEDGF(1-326), a protein devoid of heat shock elements of LEDGF, a cell survival factor, reduces P23H rhodopsin aggregates and any associated cellular damage. METHODS: ARPE-19 cells were transiently transfected/cotransfected with pLEDGF(1-326) and/or pWT-Rho (wild type)/pP23H-Rho. Rhodopsin mediated cellular damage and rescue by LEDGF(1-326) was assessed using cell viability, cell proliferation, and confocal microscopy assays. Rhodopsin monomers, oligomers, and their reduction in the presence of LEDGF(1-326) were quantified by western blot analysis. P23H rhodopsin mRNA levels in the presence and absence of LEDGF(1-326) was determined by real time quantitative PCR. PRINCIPAL FINDINGS: P23H rhodopsin reduced RPE cell viability and cell proliferation in a dose dependent manner, and disrupted the nuclear material. LEDGF(1-326) did not alter P23H rhodopsin mRNA levels, reduced its oligomers, and significantly increased RPE cell viability as well as proliferation, while reducing nuclear damage. WT rhodopsin formed oligomers, although to a smaller extent than P23H rhodopsin. Further, LEDGF(1-326) decreased WT rhodopsin aggregates. CONCLUSIONS: P23H rhodopsin as well as WT rhodopsin form aggregates in RPE cells and LEDGF(1-326) decreases these aggregates. Further, LEDGF(1-326) reduces the RPE cell damage caused by P23H rhodopsin. LEDGF(1-326) might be useful in treating cellular damage associated with protein aggregation diseases such as retinitis pigmentosa.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Fragmentos de Péptidos/metabolismo , Epitelio Pigmentado de la Retina/citología , Western Blotting , Línea Celular , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Humanos , Microscopía Confocal , Microscopía de Contraste de Fase , Fragmentos de Péptidos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Rodopsina
18.
Microvasc Res ; 82(3): 346-50, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21945644

RESUMEN

OBJECTIVE: To determine the efficacy of pazopanib eye drops in the streptozotocin induced diabetic retinopathy rat model. METHODS: A 0.5% w/v pazopanib suspension was prepared in phosphate buffered saline (PBS, pH 7.4) in the presence of 0.5% w/v sodium carboxymethyl cellulose. Brown Norway rats were divided into three groups (n=4) - (1) healthy, (2) diabetic, and (3) diabetic with treatment. The drug suspension was administered twice daily as eye drops to group 3 for 30 days. Efficacy parameters including the number of adherent leukocytes in the retinal vasculature (leukostasis), blood-retinal FITC-dextran leakage, and vitreous-to-plasma protein ratio were measured. RESULTS: Pazopanib suspension in the form of eye drops significantly reduced leukostasis (32%), FITC-dextran leakage (39%), and the vitreous-to-plasma protein ratio (64%) in diabetic animals compared to untreated diabetic group. CONCLUSION: Pazopanib eye drops can alleviate retinal complications of diabetic retinopathy.


Asunto(s)
Barrera Hematorretinal/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Retinopatía Diabética/prevención & control , Leucostasis/prevención & control , Edema Macular/prevención & control , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Sulfonamidas/farmacología , Administración Oftálmica , Animales , Glucemia/metabolismo , Proteínas Sanguíneas/metabolismo , Barrera Hematorretinal/enzimología , Peso Corporal , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/enzimología , Retinopatía Diabética/enzimología , Retinopatía Diabética/etiología , Indazoles , Leucocitos/efectos de los fármacos , Leucocitos/enzimología , Leucostasis/enzimología , Leucostasis/etiología , Edema Macular/enzimología , Edema Macular/etiología , Masculino , Terapia Molecular Dirigida , Soluciones Oftálmicas , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Tirosina Quinasas/metabolismo , Pirimidinas/administración & dosificación , Ratas , Ratas Endogámicas BN , Sulfonamidas/administración & dosificación , Factores de Tiempo , Cuerpo Vítreo/metabolismo
19.
Tuberculosis (Edinb) ; 91(1): 26-35, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21167783

RESUMEN

Mycobacterium tuberculosis (MTB) has evolved methods to evade interferon-gamma (IFNγ) mediated protection. We sought to determine the effect of MTB infection on expression of IFNγ-inducible Protein 10 (IP-10) and Monokine Induced by IFNγ (MIG), two chemokines involved in host defense. MTB infection of THP-1 cells inhibited the transcription of IP-10 and MIG. A key mechanism for the inhibition is the disruption of binding of Signal Transduction and Activation of Transcription 1-alpha (STAT1α) to its cis-regulatory element, present in the 5'-flanking region of both IP-10 and MIG promoters. Use of inhibitors specific to the nuclear factor-kappa B (NFκB) and p38 mitogen-activated protein kinase (p38(mapk)) implicate these two signaling pathways in mediating the effect of MTB on the inhibition of IFNγ-induced IP-10 and MIG mRNA expression. Interestingly, despite transcriptional inhibition, there was an unexpected increase in IP-10 and MIG protein production after combined IFNγ and MTB stimulation. MTB also inhibited IFNγ induction of MIG mRNA but augmented MIG protein in primary human monocyte-derived macrophages. The synergy between MTB and IFNγ in the induction of IP-10 and MIG protein appears to involve novel post-transcriptional events that incorporates non-canonical functions of NFκB and p38(mapk).


Asunto(s)
Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/metabolismo , Interferón gamma/metabolismo , Mycobacterium tuberculosis/genética , Tuberculosis/genética , Northern Blotting , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Activación Transcripcional/genética , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Arthritis Rheum ; 60(2): 408-18, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19180511

RESUMEN

OBJECTIVE: To examine the relative importance of tumor necrosis factor receptor I (TNFRI) signaling in the hematopoietic tissue compartment in the progression of collagen-induced arthritis (CIA), a model of rheumatoid arthritis (RA). METHODS: DBA/1 mice were administered a lethal radiation dose and were then rescued with bone marrow derived from either DBA/1 or TNFRI(-/-) mice. CIA was then induced, and disease progression was characterized. RESULTS: Surprisingly, mice with CIA that received TNFRI(-/-) donor marrow developed increased disease severity as compared with control mice with CIA. This could not be attributed to an increased primary response to collagen or to the contribution of a non-DBA genetic background. In mice that received TNFRI(-/-) bone marrow, histologic markers of advanced disease were evident shortly after initiation of the immune response to collagen and long before clinical evidence of disease. Serum TNFalpha was undetectable, whereas serum interleukin-12 p40 levels were increased, at the end point of the study in mice that received TNFRI(-/-) bone marrow. CONCLUSION: These data raise the intriguing possibility of the existence of an antiinflammatory, TNFRI-mediated circuit in the hematopoietic compartment. This circuit bears a resemblance to the switch in TNFalpha function that has been observed during the resolution of bacterial infections. These data suggest that TNFRI-mediated signals in the radioresistant tissues contribute to disease progression, whereas TNFRI-mediated signals in the radiosensitive tissues can contribute to protection from disease. We thus put forward the hypothesis that the degree of response to TNFalpha blockade in RA is dependent in part on the relative genetic strengths of these 2 pathways.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Artritis Experimental/patología , Artritis Experimental/fisiopatología , Artritis Reumatoide/patología , Artritis Reumatoide/fisiopatología , Trasplante de Médula Ósea , Resorción Ósea , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Quimera por Radiación/metabolismo , Traumatismos Experimentales por Radiación/terapia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Índice de Severidad de la Enfermedad , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA