Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Am J Pathol ; 187(9): 2008-2019, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28710903

RESUMEN

Concanavalin A (ConA) causes immune cell-mediated liver damage, but the contribution of resident nonparenchymal cells (NPCs) is also evident. Hepatic stellate cells (HSCs) induce hepatic inflammation and immunological reactions; we therefore investigated their role in ConA-induced liver injury. ConA was administered i.v. to control or HSC-depleted mice; hepatic histopathology and cytokines/chemokines were determined after 6 hours. In vitro, effects of ConA-conditioned HSC medium on hepatocytes were determined. ConA induced inflammation, sinusoidal congestion, and extensive midzonal hepatocyte death in control mice, which were strongly minimized in HSC-depleted mice. CD4 and natural killer T cells and neutrophils were markedly reduced in ConA-treated HSC-depleted mice compared with control mice. The increase in cytokines/chemokines of hepatic injury was much higher in ConA-treated control mice than in HSC-depleted mice. ConA-treated HSCs showed increased expression of interferon-ß, tumor necrosis factor-α, and CXCL1, induced oxidative stress in hepatocytes, and caused hepatocyte apoptosis. ConA induced nuclear translocation of interferon-regulatory factor-1 (IRF1) in hepatocytes in vivo, and ConA/HSC induced a similar effect in cultured hepatocytes. IRF1-knockout mice were resistant to ConA-induced liver damage, and anti-interferon ß antibody mitigated ConA/HSC-induced injury. In HSC-NPC co-culture, ConA-induced expression of inflammatory cytokines/chemokines was significantly augmented compared with NPCs alone. HSCs play an essential role in ConA-induced liver injury directly via the interferon-ß/IRF1 axis, and by modulating properties of NPCs.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Concanavalina A/toxicidad , Células Estrelladas Hepáticas/patología , Hígado/patología , Animales , Apoptosis/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones
2.
J Cell Physiol ; 231(1): 94-105, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26031389

RESUMEN

Bacterial lipopolysaccharide (LPS)-stimulated hepatic stellate cells (HSCs) produce many cytokines including IFNß, TNFα, and IL6, strongly inhibit DNA synthesis, but induce apoptosis of a small number of hepatocytes. In vivo administration of LPS (up to 10 mg/mL) causes modest inflammation and weight loss in rats but not mortality. We determined whether LPS-stimulated HSCs instigate mechanisms of hepatocyte survival. Rats received 10 mg/kg LPS (i.p.) and determinations were made at 6 h. In vitro, HSCs were treated with 100 ng/mL LPS till 24 h. The medium was transferred to hepatocytes, and determinations were made at 0-12 h. Controls were HSC-conditioned medium or medium-containing LPS. LPS treatment of rats caused autophagy in hepatocytes, a physiological process for clearance of undesirable material including injured or damaged organelles. This was accompanied by activation of c-Jun NH2 terminal kinase (JNK) and apoptosis of ~4-5% of hepatocytes. In vitro, LPS-conditioned HSC medium (LPS/HSC) induced autophagy in hepatocytes but apoptosis of only ~10% of hepatocytes. While LPS/HSC stimulated activation of JNK (associated with cell death), it also activated NFkB and ERK1/2 (associated with cell survival). LPS-stimulated HSCs produced IFNß, and LPS/HSC-induced autophagy in hepatocytes and their apoptosis were significantly inhibited by anti-IFNß antibody. Blockade of autophagy, on the other hand, strongly augmented hepatocyte apoptosis. While LPS-stimulated HSCs cause apoptosis of a subpopulation of hepatocytes by producing IFNß, they also induce cell survival mechanisms, which may be of critical importance in resistance to liver injury during endotoxemia.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Endotoxinas/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Células Estrelladas Hepáticas/citología , Hepatocitos/citología , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratas Sprague-Dawley
3.
Vet Ophthalmol ; 18(3): 242-50, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25041235

RESUMEN

OBJECTIVE: To evaluate the safety and efficacy of Pirfenidone (PFD) in the treatment of equine corneal fibrosis using an in vitro model. METHODS: Healthy donor equine corneas were collected and used to generate primary equine corneal fibroblasts (ECFs) by growing cultures in minimal essential medium supplemented with 10% fetal bovine serum. Equine corneal myofibroblasts (ECMs), used as a model of equine corneal fibrosis, were produced by growing ECF cultures in serum-free medium containing transforming growth factor ß1 (1 ng/mL). Trypan blue viability assays and changes in ECF morphology were utilized to determine the optimal PFD dose for this in vitro model. Trypan blue viability, phase-contrast microscopy, and TUNEL assays were used to evaluate the cytotoxicity of PFD. Scratch and MTT assays were used to evaluate the effect of PFD on cellular migration and proliferation. Real-time PCR, immunoblot analysis, and immunocytochemistry were employed to determine the efficacy of PFD to inhibit ECM formation in vitro. RESULTS: Topical PFD application at 200 µg/mL successfully decreased αSMA expression when compared to the TGFß1 only treatment group (P < 0.01). PFD application ≤ 200 µg/mL did not affect ECF phenotype or cellular viability and did not result in significant cytotoxicity. CONCLUSIONS: Pirfenidone safely and effectively inhibits TGFß1-induced equine corneal fibrosis in vitro. In vivo studies are warranted.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Lesiones de la Cornea/veterinaria , Enfermedades de los Caballos/tratamiento farmacológico , Piridonas/uso terapéutico , Actinas/genética , Actinas/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Córnea/citología , Lesiones de la Cornea/tratamiento farmacológico , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Caballos
4.
Am J Physiol Renal Physiol ; 307(7): F777-82, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25056353

RESUMEN

Peritoneal dialysis (PD) is a life-sustaining therapy for end-stage renal disease (ESRD), used by 10-15% of the dialysis population worldwide. Peritoneal fibrosis (PF) is a known complication of long-term PD and frequently follows episodes of peritonitis, rendering the peritoneal membrane inadequate for dialysis. Transforming growth factor (TGF)-ß is an inducer of fibrosis in several tissues and organs, and its overexpression has been correlated with PF. Animal models of peritonitis have shown an increase in expression of TGF-ß in the peritoneal tissue. Decorin, a proteoglycan and component of the extracellular matrix, inactivates TGF-ß, consequently reducing fibrosis in many tissues. Recently, gold nanoparticles (GNP) have been used for drug delivery in a variety of settings. In the present study, we tested the possibility that GNP-delivered decorin gene therapy ameliorates zymosan-mediated PF. We created a PF model using zymosan-induced peritonitis. Rats were treated with no decorin, GNP-decorin, or adeno-associated virus-decorin (AAV-decorin) and compared with controls. Tissue samples were then stained for Masson's trichrome, enface silver, and hematoxylin and eosin, and immunohistochemistry was carried out with antibodies to TGF-ß1, α-smooth muscle actin (α-SMA), and VEGF. Animals which were treated with GNP-decorin and AAV-decorin gene therapy had significant reductions in PF compared with untreated animals. Compared with untreated animals, the treated animals had better preserved peritoneal mesothelial cell size, a significant decrease in peritoneal thickness, and decreased α-SMA. Quantitative PCR measurements showed a significant decrease in the peritoneal tissue levels of α-SMA, TGF-ß, and VEGF in treated vs. untreated animals. This study shows that both GNP-delivered and AAV-mediated decorin gene therapies significantly decrease PF in vivo in a rodent model. This approach has important clinical translational potential in providing a therapeutic strategy to prevent PF in PD patients.


Asunto(s)
Decorina/genética , Terapia Genética , Fibrosis Peritoneal/prevención & control , Ratas Sprague-Dawley , Adenoviridae , Animales , Técnicas de Transferencia de Gen , Nanopartículas , Fibrosis Peritoneal/inducido químicamente , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Zimosan
5.
Vet Ophthalmol ; 17(3): 162-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23718145

RESUMEN

OBJECTIVE: To explore (i) the potential of polyethylenimine (PEI) nanoparticles as a vector for delivering genes into equine corneal fibroblasts (ECFs) using green fluorescent protein (GFP) marker gene, (ii) whether PEI nanoparticle-mediated decorin (DCN) gene therapy could be used to inhibit fibrosis in the equine cornea using an in vitro model. PROCEDURE: Polyethylenimine-DNA nanoparticles were prepared at nitrogen-to-phosphate (N-P) ratio of 15 by mixing 22 kDa linear PEI and a plasmid encoding either GFP or DCN. ECFs were generated from donor corneas as previously described. Initially, GFP was introduced into ECFs using PEI nanoparticles to confirm gene delivery, then DCN was introduced to evaluate for antifibrotic effects. GFP gene delivery was confirmed with real-time qPCR and ELISA. Changes in fibrosis after DCN therapy were quantified by measuring α-smooth muscle actin (αSMA) mRNA and protein levels with qPCR, immunostaining, and immunoblotting. Cytotoxicity was determined by evaluating cell morphology, cellular viability, and TUNEL assay. RESULTS: Polyethylenimine-green fluorescent protein-treated cultures showed 2.2 × 10(4) GFP plasmid copies/µg of cellular DNA and 2.1 pg of GFP/100 µL of lysate. PEI-DCN delivery significantly attenuated TGFß-induced transdifferentiation of fibroblasts to myofibroblasts (2-fold decrease of αSMA mRNA; P = 0.05) and significant inhibition of αSMA (49 ± 14.2%; P < 0.001) in immunocytochemical staining and immunoblotting were found. Furthermore, PEI-DNA nanoparticle delivery did not alter cellular phenotype at 24 h and cellular viability was maintained. CONCLUSIONS: Twenty-two kilo dalton Polyethylenimine nanoparticles are safe and effective for equine corneal gene therapy in vitro. PEI-mediated DCN gene delivery is effective at inhibiting TGFß-mediated fibrosis in this model.


Asunto(s)
Córnea/citología , Decorina/metabolismo , Fibroblastos/efectos de los fármacos , Técnicas de Transferencia de Gen/veterinaria , Caballos , Polietileneimina/farmacología , Animales , Células Cultivadas , Decorina/química , Decorina/genética , Fibroblastos/citología , Genes Reporteros , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Nanopartículas , Polietileneimina/química
6.
Cureus ; 16(3): e56896, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38659506

RESUMEN

Sepsis remains a formidable challenge in healthcare, characterized by a dysregulated host response to infection, leading to organ dysfunction and high mortality rates. Glutathione, a critical antioxidant and regulator of cellular redox balance, has emerged as a key player in the pathophysiology of sepsis. This comprehensive review explores the multifaceted role of glutathione in sepsis, focusing on its involvement in oxidative stress, immune modulation, and organ dysfunction. Glutathione depletion exacerbates oxidative damage and inflammatory responses, thereby contributing to the progression of sepsis. Understanding the intricate mechanisms underlying glutathione dysregulation in sepsis offers potential therapeutic avenues, with strategies targeting glutathione pathways showing promise in mitigating septic complications. However, further research is needed to optimize therapeutic approaches and identify biomarkers for patient stratification. Overall, this review underscores the importance of elucidating glutathione's role in sepsis management to improve clinical outcomes and reduce the global burden of this life-threatening condition.

7.
Ocul Surf ; 32: 13-25, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38191093

RESUMEN

PURPOSE: Corneal fibrosis and neovascularization (CNV) after ocular trauma impairs vision. This study tested therapeutic potential of tissue-targeted adeno-associated virus5 (AAV5) mediated decorin (DCN) and pigment epithelium-derived factor (PEDF) combination genes in vivo. METHODS: Corneal fibrosis and CNV were induced in New Zealand White rabbits via chemical trauma. Gene therapy in stroma was delivered 30-min after chemical-trauma via topical AAV5-DCN and AAV5-PEDF application using a cloning cylinder. Clinical eye examinations and multimodal imaging in live rabbits were performed periodically and corneal tissues were collected 9-day and 15-day post euthanasia. Histological, cellular, and molecular and apoptosis assays were used for efficacy, tolerability, and mechanistic studies. RESULTS: The AAV5-DCN and AAV5-PEDF combination gene therapy significantly reduced corneal fibrosis (p < 0.01 or p < 0.001) and CNV (p < 0.001) in therapy-given (chemical-trauma and AAV5-DCN + AAV5-PEDF) rabbit eyes compared to the no-therapy given eyes (chemical-trauma and AAV5-naked vector). Histopathological analyses demonstrated significantly reduced fibrotic α-smooth muscle actin and endothelial lectin expression in therapy-given corneas compared to no-therapy corneas on day-9 (p < 0.001) and day-15 (p < 0.001). Further, therapy-given corneas showed significantly increased Fas-ligand mRNA levels (p < 0.001) and apoptotic cell death in neovessels (p < 0.001) compared to no-therapy corneas. AAV5 delivered 2.69 × 107 copies of DCN and 2.31 × 107 copies of PEDF genes per µg of DNA. AAV5 vector and delivered DCN and PEDF genes found tolerable to the rabbit eyes and caused no significant toxicity to the cornea. CONCLUSION: The combination AAV5-DCN and AAV5-PEDF topical gene therapy effectively reduces corneal fibrosis and CNV with high tolerability in vivo in rabbits. Additional studies are warranted.


Asunto(s)
Neovascularización de la Córnea , Fibrosis , Terapia Genética , Factores de Crecimiento Nervioso , Serpinas , Animales , Conejos , Córnea/patología , Córnea/metabolismo , Neovascularización de la Córnea/terapia , Neovascularización de la Córnea/genética , Neovascularización de la Córnea/patología , Neovascularización de la Córnea/metabolismo , Decorina/genética , Decorina/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Fibrosis/terapia , Terapia Genética/métodos , Vectores Genéticos , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Serpinas/genética , Serpinas/metabolismo
8.
Mol Vis ; 18: 2598-607, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23112572

RESUMEN

PURPOSE: To explore (i) the potential of polyethylenimine (PEI)-DNA nanoparticles as a vector for delivering genes into human corneal fibroblasts, and (ii) whether the nanoparticle-mediated soluble extracellular domain of the transforming growth factor-ß type II receptor (sTGFßRII) gene therapy could be used to reduce myofibroblasts and fibrosis in the cornea using an in vitro model. METHODS: PEI-DNA nanoparticles were prepared at a nitrogen-to-phosphate ratio of 30 by mixing linear PEI and a plasmid encoding sTGFßRII conjugated to the fragment crystallizable (Fc) portion of human immunoglobulin. The PEI-DNA polyplex formation was confirmed through gel retardation assay. Human corneal fibroblasts (HCFs) were generated from donor corneas; myofibroblasts and fibrosis were induced with TGFß1 (1 ng/ml) stimulation employing serum-free conditions. The sTGFßRII conjugated to the Fc portion of human immunoglobulin gene was introduced into HCF using either PEI-DNA nanoparticles or Lipofectamine. Suitable negative and positive controls to compare selected nanoparticle and therapeutic gene efficiency were included. Delivered gene copies and mRNA (mRNA) expression were quantified with real-time quantitative PCR (qPCR) and protein with enzyme-linked immunosorbent assay (ELISA). The changes in fibrosis parameters were quantified by measuring fibrosis marker α-smooth muscle actin (SMA) mRNA and protein levels with qPCR, immunostaining, and immunoblotting. Cytotoxicity was determined using cellular viability, proliferation, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. RESULTS: PEI readily bound to plasmids to form nanoparticular polyplexes and exhibited much greater transfection efficiency (p<0.01) than the commercial reagent Lipofectamine. The PEI-DNA-treated cultures showed 4.5×10(4) plasmid copies/µg DNA in real-time qPCR and 7,030±87 pg/ml sTGFßRII protein in ELISA analyses, whereas Lipofectamine-transfected cultures demonstrated 1.9×10(3) gene copies/µg DNA and 1,640±100 pg/ml sTGFßRII protein during these assays. The PEI-mediated sTGFßRII delivery remarkably attenuated TGFß1-induced transdifferentiation of corneal fibroblasts to myofibroblasts in cultures, as indicated by threefold lower levels of SMA mRNA (p<0.01) and significant inhibition of SMA protein (up to 96±3%; p<0.001 compared to no-gene-delivered cultures) in immunocytochemical staining and immunoblotting. The nanoparticle-mediated delivery of sTGFßRII showed significantly better antifibrotic effects than the Lipofectamine under similar experimental conditions. However, the inhibition of myofibroblast in HCF cultures by sTGFßRII overexpression by either method was significantly higher than the naked vector transfection. Furthermore, PEI- or Lipofectamine-mediated sTGFßRII delivery into HCF did not alter cellular proliferation or phenotype at 12 and 24 h post-treatment. Nanoparticles treated with HCF showed more than 90% cellular viability and very low cell death (2-6 TUNEL+ cells), suggesting that the tested doses of PEI-nanoparticles do not induce significant cell death. CONCLUSIONS: This study demonstrated that PEI-DNA nanoparticles are an attractive vector for the development of nonviral corneal gene therapy approaches and that the sTGFßRII gene delivery into keratocytes could be used to control corneal fibrosis in vivo.


Asunto(s)
Córnea/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Nanopartículas/química , Polietileneimina/química , Proteínas Serina-Treonina Quinasas/farmacología , Transfección/métodos , Actinas/biosíntesis , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Córnea/patología , Fibrosis/inducido químicamente , Fibrosis/patología , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Lípidos/química , Miofibroblastos/patología , Nanopartículas/uso terapéutico , Plásmidos/química , Plásmidos/genética , Proteínas Serina-Treonina Quinasas/química , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/química , Solubilidad , Factor de Crecimiento Transformador beta1/efectos adversos
9.
J Refract Surg ; 28(4): 285-90, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22386369

RESUMEN

PURPOSE: This study investigated the efficacy and safety of vorinostat, a deacetylase (HDAC) inhibitor, in the treatment of laser-induced corneal haze following photorefractive keratectomy (PRK) in rabbits in vivo and transforming growth factor beta 1 (TGFß1) -induced corneal fibrosis in vitro. METHODS: Corneal haze in rabbits was produced with -9.00 diopters (D) PRK. Fibrosis in cultured human and rabbit corneal fibroblasts was activated with TGFß1. Vorinostat (25 µm) was topically applied once for 5 minutes on rabbit cornea immediately after PRK for in vivo studies. Vorinostat (0 to 25 µm) was given to human/rabbit corneal fibroblasts for 5 minutes or 48 hours for in vitro studies. Slit-lamp microscopy, TUNEL assay, and trypan blue were used to determined vorinostat toxicity, whereas real-time polymerase chain reaction, immunocytochemistry, and immunoblotting were used to measure its efficacy. RESULTS: Single 5-minute vorinostat (25 µm) topical application on the cornea following PRK significantly reduced corneal haze (P<.008) and fibrotic marker proteins (α-smooth muscle actin and f-actin; P<.001) without showing redness, swelling, or inflammation in rabbit eyes in vivo screened 4 weeks after PRK. Vorinostat reduced TGFß1-induced fibrosis in human and rabbit corneas in vitro in a dose-dependent manner without altering cellular viability, phenotype, or proliferation. CONCLUSIONS: Vorinostat is non-cytotoxic and safe for the eye and has potential to prevent laser-induced corneal haze in patients undergoing PRK for high myopia.


Asunto(s)
Córnea/cirugía , Opacidad de la Córnea/prevención & control , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Queratectomía Fotorrefractiva , Complicaciones Posoperatorias/prevención & control , Actinas/genética , Animales , Células Cultivadas , Córnea/efectos de los fármacos , Queratocitos de la Córnea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Fibronectinas/genética , Fibrosis/inducido químicamente , Fibrosis/prevención & control , Inhibidores de Histona Desacetilasas/efectos adversos , Humanos , Ácidos Hidroxámicos/efectos adversos , Etiquetado Corte-Fin in Situ , ARN Mensajero/metabolismo , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Crecimiento Transformador beta1/farmacología , Vorinostat
10.
Nanomedicine ; 7(4): 505-13, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21272669

RESUMEN

This study examined the gene transfer efficiency and toxicity of 2-kDa polyethylenimine conjugated to gold nanoparticles (PEI2-GNPs) in the human cornea in vitro and rabbit cornea in vivo. PEI2-GNPs with nitrogen-to-phosphorus ratios of up to 180 exhibited significant transgene delivery in the human cornea without altering the viability or phenotype of these cells. Similarly, PEI2-GNPs applied to corneal tissues collected after 12 hours, 72 hours, or 7 days exhibited appreciable gold uptake throughout the rabbit stroma with gradual clearance of GNPs over time. Transmission electron microscopy detected GNPs in the keratocytes and the extracellular matrix of the rabbit corneas. Additionally, slit-lamp biomicroscopy in live animals even 7 days after topical PEI2-GNP application to the cornea detected no inflammation, redness, or edema in rabbit eyes in vivo, with only moderate cell death and immune reactions. These results suggest that PEI2-GNPs are safe for the cornea and can potentially be useful for corneal gene therapy in vivo. FROM THE CLINICAL EDITOR: This study examined the gene transfer efficiency and toxicity of 2-kDa polyethylenimine conjugated to gold nanoparticles in the human cornea in vitro and rabbit cornea in vivo. The results suggest that PEI2-GNPs are safe for the cornea and can potentially be useful for corneal gene therapy in vivo.


Asunto(s)
Córnea/metabolismo , Oro/química , Nanopartículas del Metal/efectos adversos , Nanopartículas del Metal/química , Polietileneimina/química , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Córnea/citología , Queratocitos de la Córnea/citología , Queratocitos de la Córnea/efectos de los fármacos , Femenino , Técnicas de Transferencia de Gen , Humanos , Técnicas In Vitro , Nanotecnología/métodos , Conejos
11.
Mol Vis ; 16: 2494-501, 2010 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-21139995

RESUMEN

PURPOSE: This study tested whether controlled drying of the cornea increases vector absorption in mouse and rabbit corneas in vivo and human cornea ex vivo, and studied the effects of corneal drying on gene transfer, structure and inflammatory reaction in the mouse cornea in vivo. METHODS: Female C57 black mice and New Zealand White rabbits were used for in vivo studies. Donor human corneas were used for ex vivo experiments. A hair dryer was used for drying the corneas after removing corneal epithelium by gentle scraping. The corneas received no, once, twice, thrice, or five times warm air for 10 s with a 5 s interval after each 10 s hair dryer application. Thereafter, balanced salt solution (BSS) was topically applied immediately on the cornea for 2 min using a custom-cloning cylinder. The absorbed BSS was quantified using Hamilton microsyringes. The adeno-associated virus 8 (AAV8) vector (1.1×10(8) genomic copies/µl) expressing marker gene was used to study the effect of corneal drying on gene transfer. Animals were sacrificed on day 14 and gene expression was analyzed using commercial staining kit. Morphological changes and infiltration of inflammatory cells were examined with H & E staining and immunocytochemistry. RESULTS: Mice, rabbit or human corneas subjected to no or 10 s drying showed 6%-8% BSS absorption whereas 20, 30, or 50 s corneal drying showed significantly high 14%-19% (p<0.001), 21%-22% (p<0.001), and 25%-27% (p<0.001) BSS absorption, respectively. The AAV8 application on mouse cornea after 50 s drying showed significantly higher transgene delivery (p<0.05) in vivo with mild-to-moderate changes in corneal morphology. The 30 s of drying also showed significantly (p<0.05) high transgene delivery in mouse stroma in vivo without jeopardizing corneal morphology whereas 10 or 20 s drying showed moderate degree of gene transfer with no altered corneal morphology. Corneas that underwent 50 s drying showed high CD11b-positive cells (p<0.01) compared to control corneas whereas 20 or 30 s air-dried corneas showed insignificant CD11b-positive cells compared to control corneas. CONCLUSIONS: Controlled corneal drying with hair dryer increases vector absorption significantly. The dispensing of efficacious AAV serotype into cornea with optimized minimally invasive topical application technique could provide high and targeted expression of therapeutic genes in the stroma in vivo without causing significant side effects.


Asunto(s)
Córnea/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Absorción , Administración Tópica , Fosfatasa Alcalina/metabolismo , Animales , Antígeno CD11b/metabolismo , Femenino , Expresión Génica , Vectores Genéticos/administración & dosificación , Humanos , Inmunohistoquímica , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Conejos , Soluciones
12.
Immunol Invest ; 38(8): 749-61, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19860586

RESUMEN

Crude antigenic preparations from heterologous filarial parasites gave false positive results because of complex nature of these antigens and their cross-reactivity with other helminth parasites. In the present study, efforts have been made to isolate and characterize the antigens from Setaria cervi important for diagnostic purposes. The fractionation of S. cervi somatic antigenic preparation on Sephacryl S-200 resulted in separation of three major antigenic peak fractions. Crossed immunoelectrophoretic analysis, using immune rabbit serum, revealed 13-14 antigens in SFP-I pool fraction, which showed high reactivity with filarial patients sera as compared to other two pool fractions. This SFP-I fraction was further purified by DEAE-Cellulose column chromatography. Out of the 4 antigen pool fractions, DFP-IV fraction showed high ELISA reactivity with filarial patient serum pool (Wuchereria bancrofti and Brugia malayi) as compared to other fractions. The SDS-PAGE analysis of DFP-IV fraction revealed 2 major and 1 minor protein bands (mol. wt. range 65-70 kDa). Crossed immunoelectrophoresis also showed the presence of 3 antigenic peaks in DFP-IV fraction. The purified DFP-IV fraction showed high reactivity with filarial patients sera but did not cross-react with sera from ascaris and hookworm infections thereby suggesting the filaria-specificity and potential for immunodiagnosis of human filariasis.


Asunto(s)
Antígenos Helmínticos/inmunología , Brugia Malayi/inmunología , Pruebas Inmunológicas , Setaria (Nematodo)/inmunología , Setariasis/inmunología , Wuchereria bancrofti/inmunología , Animales , Antígenos Helmínticos/análisis , Antígenos Helmínticos/aislamiento & purificación , Brugia Malayi/parasitología , Fraccionamiento Celular , Cromatografía DEAE-Celulosa , Mezclas Complejas/inmunología , Reacciones Cruzadas , Humanos , Sueros Inmunes , Estadios del Ciclo de Vida , Conejos , Setaria (Nematodo)/crecimiento & desarrollo , Setariasis/diagnóstico , Setariasis/parasitología , Wuchereria bancrofti/parasitología
13.
Acta Parasitol ; 62(1): 29-37, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28030352

RESUMEN

Filarial parasites are complex mixtures of antigenic proteins and characterization of these antigenic molecules is essential to identify the diagnostically important filaria-specific antigens. In the present study, we have fractionated the somatic extracts from adults of Setaria cervi (bovine filarial parasite) on preparative SDS-polyacrylamide gel electrophoresis and tested the immunoreactivity of the separated gel fractions with polyclonal antibodies against filarial excretory-secretory antigens as well as filarial patients sera. The SDS-PAGE analysis of gel eluted fractions revealed 1 protein band in F-1 fraction, 2 protein bands in F-2 fraction and 2-3 protein bands in all other fractions (F3- F11). Seven gel eluted fractions (F1, F2, F3, F4, F5, F6 and F11) showed high ELISA reactivity with the polyclonal antibody (against excretory-secretory antigen) and four of these fractions (F-1, F-2, F3 and F6) exhibited high ELISA reactivity with antibodies present in filarial patient sera. The reactivities of the gel fractions (F1 and F2), recognized by filarial patients sera, were also tested with the monoclonal antibody (detecting the filarial circulating antigen). The F1 and F2 gel eluted fractions were found to have the target antigen of monoclonal antibody as evident by high reactivity with the monoclonal antibody in ELISA and immunoblotting. The S. cervi gel eluted F1 fraction (containing single antigen) could detect antibodies in filarial patients sera and not in non-filarial sera thereby suggesting its usefulness for specific serodiagnosis of human filariasis.


Asunto(s)
Antígenos Helmínticos/inmunología , Electroforesis en Gel de Poliacrilamida , Proteínas del Helminto/metabolismo , Setaria (Nematodo)/metabolismo , Animales , Anticuerpos Antihelmínticos , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Proteínas del Helminto/genética , Humanos , Sueros Inmunes , Pruebas Inmunológicas , Masculino , Ratones , Conejos , Setariasis/sangre , Setariasis/inmunología
14.
PLoS One ; 8(12): e82159, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24349206

RESUMEN

With their location in the perisinusoidal space of Disse, hepatic stellate cells (HSCs) communicate with all of the liver cell types both by physical association (cell body as well as cytosolic processes penetrating into sinusoids through the endothelial fenestrations) and by producing several cytokines and chemokines. Bacterial lipopolysaccharide (LPS), circulating levels of which are elevated in liver diseases and transplantation, stimulates HSCs to produce increased amounts of cytokines and chemokines. Although recent research provides strong evidence for the role of HSCs in hepatic inflammation and immune regulation, the number of HSC-elaborated inflammatory and immune regulatory molecules may be much greater then known at the present time. Here we report time-dependent changes in the gene expression profile of inflammatory and immune-regulatory molecules in LPS-stimulated rat HSCs, and their validation by biochemical analyses. LPS strongly up-regulated LPS-response elements (TLR2 and TLR7) but did not affect TLR4 and down-regulated TLR9. LPS also up-regulated genes in the MAPK, NFκB, STAT, SOCS, IRAK and interferon signaling pathways, numerous CC and CXC chemokines and IL17F. Interestingly, LPS modulated genes related to TGFß and HSC activation in a manner that would limit their activation and fibrogenic activity. The data indicate that LPS-stimulated HSCs become a major cell type in regulating hepatic inflammatory and immunological responses by altering expression of numerous relevant genes, and thus play a prominent role in hepatic pathophysiology including liver diseases and transplantation.


Asunto(s)
Células Estrelladas Hepáticas/inmunología , Células Estrelladas Hepáticas/patología , Inflamación/genética , Lipopolisacáridos/farmacología , Hígado/inmunología , Hígado/patología , Transcriptoma/genética , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/genética , Quimiocinas/genética , Quimiocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/enzimología , Inflamación/inmunología , Inflamación/patología , Hígado/metabolismo , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Elementos de Respuesta/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
15.
PLoS One ; 8(6): e66434, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23799103

RESUMEN

This study examined the effects of BMP7 gene transfer on corneal wound healing and fibrosis inhibition in vivo using a rabbit model. Corneal haze in rabbits was produced with the excimer laser performing -9 diopters photorefractive keratectomy. BMP7 gene was introduced into rabbit keratocytes by polyethylimine-conjugated gold nanoparticles (PEI2-GNPs) transfection solution single 5-minute topical application on the eye. Corneal haze and ocular health in live animals was gauged with stereo- and slit-lamp biomicroscopy. The levels of fibrosis [α-smooth muscle actin (αSMA), F-actin and fibronectin], immune reaction (CD11b and F4/80), keratocyte apoptosis (TUNEL), calcification (alizarin red, vonKossa and osteocalcin), and delivered-BMP7 gene expression in corneal tissues were quantified with immunofluorescence, western blotting and/or real-time PCR. Human corneal fibroblasts (HCF) and in vitro experiments were used to characterize the molecular mechanism mediating BMP7's anti-fibrosis effects. PEI2-GNPs showed substantial BMP7 gene delivery into rabbit keratocytes in vivo (2×10(4) gene copies/ug DNA). Localized BMP7 gene therapy showed a significant corneal haze decrease (1.68±0.31 compared to 3.2±0.43 in control corneas; p<0.05) in Fantes grading scale. Immunostaining and immunoblot analyses detected significantly reduced levels of αSMA (46±5% p<0.001) and fibronectin proteins (48±5% p<0.01). TUNEL, CD11b, and F4/80 assays revealed that BMP7 gene therapy is nonimmunogenic and nontoxic for the cornea. Furthermore, alizarin red, vonKossa and osteocalcin analyses revealed that localized PEI2-GNP-mediated BMP7 gene transfer in rabbit cornea does not cause calcification or osteoblast recruitment. Immunofluorescence of BMP7-transefected HCFs showed significantly increased pSmad-1/5/8 nuclear localization (>88%; p<0.0001), and immunoblotting of BMP7-transefected HCFs grown in the presence of TGFß demonstrated significantly enhanced pSmad-1/5/8 (95%; p<0.001) and Smad6 (53%, p<0.001), and decreased αSMA (78%; p<0.001) protein levels. These results suggest that localized BMP7 gene delivery in rabbit cornea modulates wound healing and inhibits fibrosis in vivo by counter balancing TGFß1-mediated profibrotic Smad signaling.


Asunto(s)
Proteína Morfogenética Ósea 7/genética , Enfermedades de la Córnea/terapia , Sustancia Propia/patología , Animales , Proteína Morfogenética Ósea 7/biosíntesis , Células Cultivadas , Córnea , Enfermedades de la Córnea/patología , Sustancia Propia/metabolismo , Femenino , Fibroblastos/metabolismo , Fibrosis , Dosificación de Gen , Terapia Genética , Oro , Humanos , Nanopartículas del Metal , Conejos , Transducción de Señal , Proteínas Smad/metabolismo , Transfección , Resultado del Tratamiento
16.
Prog Retin Eye Res ; 31(1): 43-64, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21967960

RESUMEN

Successful restoration of vision in human patients with gene therapy affirmed its promise to cure ocular diseases and disorders. The efficacy of gene therapy is contingent upon vector and mode of therapeutic DNA introduction into targeted cells/tissues. The cornea is an ideal tissue for gene therapy due to its ease of access and relative immune-privilege. Considerable progress has been made in the field of corneal gene therapy in last 5 years. Several new gene transfer vectors, techniques and approaches have evolved. Although corneal gene therapy is still in its early stages of development, the potential of gene-based interventions to treat corneal abnormalities has begun to surface. Identification of next generation viral and nanoparticle vectors, characterization of delivered gene levels, localization, and duration in the cornea, and significant success in controlling corneal disorders, particularly fibrosis and angiogenesis, in experimental animal disease models, with no major side effects have propelled gene therapy a step closer toward establishing gene-based therapies for corneal blindness. Recently, researchers have assessed the delivery of therapeutic genes for corneal diseases and disorders due to trauma, infections, chemical, mechanical, and surgical injury, and/or abnormal wound healing. This review provides an update on the developments in gene therapy for corneal diseases and discusses the barriers that hinder its utilization for delivering genes in the cornea.


Asunto(s)
Enfermedades de la Córnea/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Humanos
17.
Invest Ophthalmol Vis Sci ; 52(7): 4833-41, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21551414

RESUMEN

PURPOSE: This study tested a hypothesis that tissue-selective targeted decorin gene therapy delivered to the stroma with adeno-associated virus serotype 5 (AAV5) inhibits corneal fibrosis in vivo without significant side effects. METHODS: An in vivo rabbit model of corneal fibrosis was used. Targeted decorin gene therapy was delivered to the rabbit cornea by a single topical application of AAV5 (100 µL; 6.5 × 10(12) µg/mL) onto the bare stroma for 2 minutes. The levels of corneal fibrosis were determined with stereomicroscopy, slit lamp biomicroscopy, α-smooth muscle actin (αSMA), fibronectin, and F-actin immunocytochemistry, and/or immunoblotting. CD11b, F4/80 immunocytochemistry, and TUNEL assay were used to examine immunogenicity and cytotoxicity of AAV5 to the cornea. Transmission electron microscopy (TEM) was used to investigate ultrastructural features. Slot-blot-quantified the copy number of AAV5-delivered decorin genes. RESULTS: Selective decorin delivery into the stroma showed a significant (P < 0.01) decrease in corneal haze (1.3 ± 0.3) compared with the no-decorin-delivered control rabbit corneas (3 ± 0.4) quantified using slit lamp biomicroscopy. Immunostaining and immunoblot analyses detected significantly reduced levels of αSMA, F-actin, and fibronectin proteins (59%-73%; P < 0.001 or <0.01) in decorin-delivered rabbit corneas compared with the no-decorin-delivered controls. The visual clinical eye examination, slit lamp clinical studies, TUNEL, CD11b, and F4/80 assays revealed that AAV5-mediated decorin gene therapy is nonimmunogenic and nontoxic for the cornea. TEM studies suggested that decorin gene delivery does not jeopardize collagen fibrillogenesis as no significant differences in collagen fibril diameter and arrangement were observed in decorin-delivered and no-decorin-delivered control corneas. CONCLUSIONS: Tissue-targeted AAV5-mediated decorin gene therapy is effective and safe for treating corneal fibrosis in vivo.


Asunto(s)
Cicatriz/terapia , Córnea/ultraestructura , Opacidad de la Córnea/prevención & control , Decorina/administración & dosificación , Terapia Genética/métodos , Administración Tópica , Animales , Apoptosis , Cicatriz/etiología , Cicatriz/patología , Opacidad de la Córnea/etiología , Opacidad de la Córnea/patología , Dependovirus , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Immunoblotting , Etiquetado Corte-Fin in Situ , Microscopía Electrónica de Transmisión de Rastreo , Plásmidos , Conejos
18.
PLoS One ; 6(10): e26432, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22039486

RESUMEN

Decorin, small leucine-rich proteoglycan, has been shown to modulate angiogenesis in nonocular tissues. This study tested a hypothesis that tissue-selective targeted decorin gene therapy delivered to the rabbit stroma with adeno-associated virus serotype 5 (AAV5) impedes corneal neovascularization (CNV) in vivo without significant side effects. An established rabbit CNV model was used. Targeted decorin gene therapy in the rabbit stroma was delivered with a single topical AAV5 titer (100 µl; 5×10(12) vg/ml) application onto the stroma for two minutes after removing corneal epithelium. The levels of CNV were examined with stereomicroscopy, H&E staining, lectin, collagen type IV, CD31 immunocytochemistry and CD31 immunoblotting. Real-time PCR quantified mRNA expression of pro- and anti-angiogenic genes. Corneal health in live animals was monitored with clinical, slit-lamp and optical coherence tomography biomicroscopic examinations. Selective decorin delivery into stroma showed significant 52% (p<0.05), 66% (p<0.001), and 63% (p<0.01) reduction at early (day 5), mid (day 10), and late (day 14) stages of CNV in decorin-delivered rabbit corneas compared to control (no decorin delivered) corneas in morphometric analysis. The H&E staining, lectin, collagen type IV, CD31 immunostaining (57-65, p<0.5), and CD31 immunoblotting (62-67%, p<0.05) supported morphometric findings. Quantitative PCR studies demonstrated decorin gene therapy down-regulated expression of VEGF, MCP1 and angiopoietin (pro-angiogenic) and up-regulated PEDF (anti-angiogenic) genes. The clinical, biomicroscopy and transmission electron microscopy studies revealed that AAV5-mediated decorin gene therapy is safe for the cornea. Tissue-targeted AAV5-mediated decorin gene therapy decreases CNV with no major side effects, and could potentially be used for treating patients.


Asunto(s)
Córnea/irrigación sanguínea , Decorina/uso terapéutico , Dependovirus/genética , Marcación de Gen , Terapia Genética , Neovascularización Patológica/terapia , Animales , Secuencia de Bases , Western Blotting , Cartilla de ADN , Decorina/genética , Femenino , Inmunohistoquímica , ARN Mensajero/genética , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Tomografía de Coherencia Óptica
19.
PLoS One ; 6(4): e18771, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21533273

RESUMEN

Untargeted and uncontrolled gene delivery is a major cause of gene therapy failure. This study aimed to define efficient and safe tissue-selective targeted gene therapy approaches for delivering genes into keratocytes of the cornea in vivo using a normal or diseased rabbit model. New Zealand White rabbits, adeno-associated virus serotype 5 (AAV5), and a minimally invasive hair-dryer based vector-delivery technique were used. Fifty microliters of AAV5 titer (6.5×10(12) vg/ml) expressing green fluorescent protein gene (GFP) was topically applied onto normal or diseased (fibrotic or neovascularized) rabbit corneas for 2-minutes with a custom vector-delivery technique. Corneal fibrosis and neovascularization in rabbit eyes were induced with photorefractive keratectomy using excimer laser and VEGF (630 ng) using micropocket assay, respectively. Slit-lamp biomicroscopy and immunocytochemistry were used to confirm fibrosis and neovascularization in rabbit corneas. The levels, location and duration of delivered-GFP gene expression in the rabbit stroma were measured with immunocytochemistry and/or western blotting. Slot-blot measured delivered-GFP gene copy number. Confocal microscopy performed in whole-mounts of cornea and thick corneal sections determined geometric and spatial localization of delivered-GFP in three-dimensional arrangement. AAV5 toxicity and safety were evaluated with clinical eye exam, stereomicroscopy, slit-lamp biomicroscopy, and H&E staining. A single 2-minute AAV5 topical application via custom delivery-technique efficiently and selectively transduced keratocytes in the anterior stroma of normal and diseased rabbit corneas as evident from immunocytochemistry and confocal microscopy. Transgene expression was first detected at day 3, peaked at day 7, and was maintained up to 16 weeks (longest tested time point). Clinical and slit-lamp eye examination in live rabbits and H&E staining did not reveal any significant changes between AAV5-treated and untreated control corneas. These findings suggest that defined gene therapy approaches are safe for delivering genes into keratocytes in vivo and has potential for treating corneal disorders in human patients.


Asunto(s)
Enfermedades de la Córnea/terapia , Terapia Genética , Animales , Enfermedades de la Córnea/cirugía , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Microscopía Confocal , Queratectomía Fotorrefractiva , Conejos , Factor A de Crecimiento Endotelial Vascular/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA