Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Eur J Haematol ; 106(2): 230-240, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33107092

RESUMEN

Circulating cell-free DNA (cfDNA) has the potential to capture spatial genetic heterogeneity in myeloma (MM) patients. We assessed whether cfDNA levels vary according to risk status defined by the 70 gene expression profile (GEP70). cfDNA levels in 77 patients were significantly higher in the GEP70 high-risk (HR) group compared to the low-risk (LR) group and correlated weakly with clinical markers including lactate dehydrogenase, ß2 -microglobulin, and ISS. Patients with high cfDNA levels were associated with a worse PFS (hazard ratio 6.4; 95% CI of ratio 1.9-22) and OS (hazard ratio 4.4; 95% CI of ratio 1.2-15.7). Circulating tumor DNA (ctDNA) was elevated in the HR group and ctDNA correlated strongly with GEP70 risk score (Spearman r = .69, P = .0027). cfDNA concentrations were significantly elevated between days 3-5 after chemotherapy before falling back to baseline levels. ctDNA in two patients showed a similar spike in levels between days 3 and 5 after chemotherapy with a concomitant increase in allele fraction of KRAS mutations. We assessed cfDNA levels in 25 patients with smoldering myeloma with serial samples and showed increased allele fraction of mutated KRAS at progression in cfDNA. Our study shows that cfDNA is a dynamic tool to capture genetic events in myeloma.


Asunto(s)
Biomarcadores de Tumor , ADN Tumoral Circulante , Mieloma Múltiple/genética , Alelos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Médula Ósea , Biología Computacional/métodos , Progresión de la Enfermedad , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Biopsia Líquida , Mieloma Múltiple/sangre , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/terapia , Pronóstico , Mieloma Múltiple Quiescente/sangre , Mieloma Múltiple Quiescente/diagnóstico , Mieloma Múltiple Quiescente/genética , Mieloma Múltiple Quiescente/terapia , Resultado del Tratamiento , Carga Tumoral , Proteínas ras/genética
2.
Haematologica ; 105(4): 1055-1066, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31221783

RESUMEN

MYC is a widely acting transcription factor and its deregulation is a crucial event in many human cancers. MYC is important biologically and clinically in multiple myeloma, but the mechanisms underlying its dysregulation are poorly understood. We show that MYC rearrangements are present in 36.0% of newly diagnosed myeloma patients, as detected in the largest set of next generation sequencing data to date (n=1,267). Rearrangements were complex and associated with increased expression of MYC and PVT1, but not other genes at 8q24. The highest effect on gene expression was detected in cases where the MYC locus is juxtaposed next to super-enhancers associated with genes such as IGH, IGK, IGL, TXNDC5/BMP6, FAM46C and FOXO3 We identified three hotspots of recombination at 8q24, one of which is enriched for IGH-MYC translocations. Breakpoint analysis indicates primary myeloma rearrangements involving the IGH locus occur through non-homologous end joining, whereas secondary MYC rearrangements occur through microhomology-mediated end joining. This mechanism is different to lymphomas, where non-homologous end joining generates MYC rearrangements. Rearrangements resulted in overexpression of key genes and chromatin immunoprecipitation-sequencing identified that HK2, a member of the glucose metabolism pathway, is directly over-expressed through binding of MYC at its promoter.


Asunto(s)
Genes myc , Mieloma Múltiple , ARN Largo no Codificante/genética , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Humanos , Hibridación Fluorescente in Situ , Mieloma Múltiple/genética , Proteína Disulfuro Isomerasas , Translocación Genética
3.
Exp Eye Res ; 127: 143-52, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25088354

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of irreversible blindness. This study was done to characterize dry AMD-like changes in mouse retinal pigment epithelium (RPE) and retina after polyethylene glycol (PEG) treatment. We injected male C57BL/6 mice subretinally with PBS, 0.025, 0.25, 0.5 and 1.0 mg of PEG-400 and the animals were sacrificed on day 5. Eyes were harvested and processed for histological analysis. In all other experiments 0.5 mg PEG was injected and animals were sacrificed on days 1, 3, 5 or 14. Paraffin, 5 µm and plastic, 1 µm and 80 nm sections were used for further analysis. Subretinal injection of 0.5 mg PEG induced a 32% reduction of outer nuclear layer (ONL) thickness, 61% decrease of photoreceptor outer and inner segment length, 49% decrease of nuclear density in the ONL and 31% increase of RPE cell density by day 5 after injection. The maximum level of TUNEL positive nuclei in the ONL (6.8 + 1.99%) was detected at day 5 after PEG injection and co-localized with Casp3act. Histological signs of apoptosis were observed in the ONL by light or electron microscopy. Degeneration of RPE cells was found in PEG injected eyes. Gene expression data identified several genes reported to be involved in human AMD. C3, Cfi, Serping1, Mmp9, Htra1 and Lpl were up-regulated in PEG injected eyes compared to PBS controls. PEG leads to morphological and gene expression changes in RPE and retina consistent with dry AMD. This model will be useful to investigate dry AMD pathogenesis and treatment.


Asunto(s)
Modelos Animales de Enfermedad , Atrofia Geográfica/patología , Células Fotorreceptoras de Vertebrados/patología , Polietilenglicoles/toxicidad , Epitelio Pigmentado de la Retina/patología , Aminopeptidasas/genética , Animales , Apoptosis , Autofagia , Proteína Inhibidora del Complemento C1/genética , Complemento C3/genética , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/fisiología , Atrofia Geográfica/inducido químicamente , Atrofia Geográfica/genética , Serina Peptidasa A1 que Requiere Temperaturas Altas , Etiquetado Corte-Fin in Situ , Inyecciones Intraoculares , Masculino , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Serina Endopeptidasas/genética , Factores de Tiempo
4.
Biochim Biophys Acta ; 1823(8): 1264-72, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22633972

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of central blindness in the elderly population. The wet type of AMD is characterized by extensive growth of new vessels. One of the effective strategies to treat wet AMD is to limit the choroidal neovascularization (CNV). We studied the effects of adiponectin peptide I (APNpI) on new vessel growth in laser-induced rat model of wet AMD and on rat choroidal endothelial cell (CEC) culture. CNV size and vessel density were investigated by microscopy. Immunohistochemical staining (IHC) for von Willebrand Factor (vWF), APN, APN receptors 1 (AdipoR1), 2 (AdipoR2), VEGF, VEGF receptor 2 (VEGF-R2), proliferating cell nuclear antigen (PCNA) was performed in CNV area. The mRNA expression of VEGF and VEGF-R2 in RPE-choroid was investigated by RT-PCR and real-time PCR. APNpI inhibited area of CNV by 4 fold, number of vWF positive vessels by 99% and area of subretinal tissue by 40%. The expression of VEGF and VEGF-R2 at mRNA and protein levels decreased after APNpI treatment in vivo. Proliferative index (PCNA) was 5 folds less in laser spots of APNpI treated rats compared to controls. In conclusion, APNpI inhibited formation of new vessels in rat model of CNV by decreasing VEGF, VEGF-R2 expression and cell proliferation. Thus, APNpI may have potential therapeutic use for AMD treatment since it significantly inhibited CNV.


Asunto(s)
Adiponectina/farmacología , Coroides/irrigación sanguínea , Neovascularización Coroidal/prevención & control , Fragmentos de Péptidos/farmacología , Degeneración Macular Húmeda/tratamiento farmacológico , Adiponectina/uso terapéutico , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Coroides/efectos de los fármacos , Coroides/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Expresión Génica/efectos de los fármacos , Masculino , Fragmentos de Péptidos/uso terapéutico , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas Endogámicas BN , Receptores de Adiponectina/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
J Biol Chem ; 286(18): 16229-37, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21454496

RESUMEN

In this study, we describe a new method for inducing choroidal neovascularization (CNV) in C57BL/6 mice, an animal model of wet age-related macular degeneration (AMD). AMD is a disease that causes central blindness in humans. We injected PEG-8 subretinally in different doses (0.125-2 mg) to induce CNV. After PEG-8 injection, we examined CNV at several time points (days 3-42). We also used Western blotting, immunohistochemistry, and ELISA to examine the complement component C3 split products, C9, VEGF, TGF-ß2, and basic FGF. As early as day 1 after treatment, we found that a single subretinal injection of 1 mg of PEG-8 increased the C3 split products and the C9, TGF-ß2, and basic FGF levels in the retinal pigment epithelium-choroid tissue. By day 3 after PEG-8 injection, the intraocular activation of the complement system caused induction and progression of CNV, including new vessels penetrating the Bruch's membrane. At day 5 after PEG-8 injection, we observed a fully developed CNV and retinal degeneration. Thus, in this study, we present a new, inexpensive, and accelerated mouse model of CNV that may be useful to study AMD.


Asunto(s)
Neovascularización Coroidal/inducido químicamente , Modelos Animales de Enfermedad , Portadores de Fármacos/efectos adversos , Degeneración Macular , Polietilenglicoles/efectos adversos , Animales , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Complemento C3/metabolismo , Complemento C9/metabolismo , Portadores de Fármacos/farmacología , Ratones , Polietilenglicoles/farmacología , Factores de Tiempo , Factor de Crecimiento Transformador beta2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Biol Chem ; 286(23): 20991-1001, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21515678

RESUMEN

The present study investigated the interactions among the complement membrane attack complex (MAC), CCL2, and VEGF that occur in vivo during the development of choroidal neovascularization (CNV). We first investigated the sequential expression of MAC, CCL2, and VEGF during laser-induced CNV in C57BL/6 mice. Increased MAC deposition was detected at 1 h, CCL2 increased at 3 h, and VEGF was up-regulated at day 3 post-laser treatment. These results suggested that during laser-induced CNV, MAC, CCL2 and VEGF are formed and/or expressed in the following order: MAC → CCL2 → VEGF. To determine the cross-talk between MAC, CCL2, and VEGF during laser-induced CNV, neutralizing antibodies were injected both systemically and locally to block the bioactivity of each molecule. Blocking MAC formation inhibited CCL2 and VEGF expression and also limited CNV formation, whereas neutralization of CCL2 bioactivity did not affect MAC deposition; however, it reduced VEGF expression and CNV formation. When bioactivity of VEGF was blocked, CNV formation was significantly inhibited, but MAC deposition was not affected. Together, our results demonstrate that MAC is an upstream mediator and effect of MAC on the development of laser-induced CNV can be attributed to its direct effect on VEGF as well as its effect on VEGF that is mediated by CCL2. Understanding the interplay between immune mediators is critical to gain insight into the pathogenesis of CNV.


Asunto(s)
Quimiocina CCL2/biosíntesis , Neovascularización Coroidal/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Regulación de la Expresión Génica , Rayos Láser/efectos adversos , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Anticuerpos Neutralizantes/farmacología , Neovascularización Coroidal/etiología , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Ratones , Factores de Tiempo
7.
Retina ; 32(6): 1171-80, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22088983

RESUMEN

PURPOSE: The purpose of this article is to evaluate the effect of nicotine on anti-vascular endothelial growth factor therapy in the treatment of neovascular age-related macular degeneration. METHODS: One group of mice received nicotine in drinking water and the other group received water only. Choroidal neovascularization (CNV) was induced with a laser. Nicotinic acetylcholine receptor-α7 (nAChRα7) expression was evaluated by immunohistochemistry. Bevacizumab or adiponectin peptide II (APNpII) was injected intravitreally on Day 7 postlaser, and the effects were evaluated on Days 14 and 21. α-Bungarotoxin was injected intraperitoneally on Days 2 to 5, and its effect was evaluated on Day 14. RESULTS: Expression of nAChRα7 was 2 to 7 times higher between Days 3 and 7 postlaser compared with naive mice. In water-fed mice, APNpII, bevacizumab, and α-bungarotoxin significantly reduced CNV size. In nicotine-fed mice, treatment with APNpII or bevacizumab did not significantly reduce CNV size, whereas α-bungarotoxin did have an effect. Comparing water- and nicotine-fed mice, CNV size was 61% to 86% smaller in water-fed mice except for the α-bungarotoxin group, where there was no difference. Platelet-derived growth factor and vascular endothelial growth factor expression was 1.5- to 2.5-fold higher at Day 14 in nicotine-treated mice. CONCLUSION: Nicotine significantly blocks the effect of anti-vascular endothelial growth factor therapy in the treatment of laser-induced neovascular age-related macular degeneration. nAChRα7 is significantly upregulated during the formation of CNV, and treatment with an nAChRα7 antagonist decreases CNV size irrespective of nicotine administration.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Nicotina/farmacología , Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adiponectina/uso terapéutico , Análisis de Varianza , Animales , Bevacizumab , Bungarotoxinas/farmacología , Coroides/metabolismo , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Nicotínicos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7
8.
J Biol Chem ; 285(44): 33826-33, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20736175

RESUMEN

This study was designed to explore the effect of recombinant, membrane-targeted CD59 (rCD59-APT542) on the growth and size of fully developed neovascular complex using the murine model of laser-induced choroidal neovascularization (CNV). CNV was induced by laser photocoagulation in C57BL/6 mice using an argon laser, and the animals received rCD59-APT542 via intravitreal (ivt) route. Western blot analysis, immunohistochemistry, and total complement hemolytic assay demonstrated that exogenously administered rCD59-APT542 was incorporated as well as retained in RPE and choroid and was functionally active in vivo. Single ivt injection during the growth of the CNV (i.e. at day 3 post-laser) resulted in ∼79% inhibition of the further growth of neovascular complex. The size of the CNV complex was significantly (p < 0.05) reduced by the administration of rCD59-APT542 after the CNV complex has fully developed (i.e. at day 7 post-laser). Treatment with rCD59-APT542 blocked the formation of membrane attack complex (MAC), increased apoptosis and decreased cell proliferation in the neovascular complex. On the basis of results presented here we conclude that recombinant membrane targeted CD59 inhibited the growth of the CNV complex and reduced the size of fully developed CNV in the laser-induced mouse model. We propose that a combination of two mechanisms: increased apoptosis and decreased cell proliferation, both resulting from local inhibition of MAC, may be responsible for inhibition of CNV by rCD59-APT542.


Asunto(s)
Antígenos CD59/metabolismo , Neovascularización Coroidal/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Recombinantes/química , Animales , Apoptosis , Membrana Celular/metabolismo , Proliferación Celular , Proteínas del Sistema Complemento/química , Inmunohistoquímica , Inflamación , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Am J Pathol ; 177(4): 1870-80, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20813971

RESUMEN

The objective of this study was to explore the relationship between local (ie, ocular) complement factor H (CFH) and choroidal neovascularization (CNV) associated with wet age-related macular degeneration (AMD), a leading cause of irreversible blindness, in laser-treated C57BL/6 mice. Immunohistochemical and RT-PCR analysis of retinal pigmented epithelium (RPE)-choroid sclera revealed that the expression of CFH was down-regulated on day 1 with a dramatic increase on days 5 and 7 postlaser injury. Flat mount and Western blot analysis further revealed that membrane attack complex (MAC) expression was up-regulated on days 1 and 3 postlaser injury; however, MAC was down-regulated on days 5 and 7 postinjury but was still higher than in non-injured mice. Similar patterns for CFH and MAC were observed for RPE cells when serial paraffin sections of the laser spots were analyzed. Subretinal injection of siRNA directed against CFH resulted in a threefold suppression of CFH in the RPE and choroid without affecting either CFH levels in the liver or the functional activity of the alternative pathway in the peripheral blood. Ocular knock-down of CFH resulted in increased MAC deposition, which leads to the early onset as well as exacerbation of laser-induced CNV. In conclusion, our findings provide evidence that CFH present on RPE and choroid regulates local MAC formation that is critical for the development of laser-induced CNV.


Asunto(s)
Neovascularización Coroidal/etiología , Neovascularización Coroidal/metabolismo , Factor H de Complemento/fisiología , Modelos Animales de Enfermedad , Rayos Láser/efectos adversos , Epitelio Pigmentado Ocular/metabolismo , Animales , Western Blotting , Neovascularización Coroidal/patología , Factor H de Complemento/antagonistas & inhibidores , Técnicas para Inmunoenzimas , Degeneración Macular/etiología , Degeneración Macular/metabolismo , Degeneración Macular/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Epitelio Pigmentado Ocular/patología , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Visión Ocular/fisiología
10.
Clin Cancer Res ; 26(10): 2422-2432, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31988198

RESUMEN

PURPOSE: Copy-number changes and translocations have been studied extensively in many datasets with long-term follow-up. The impact of mutations remains debated given the short time to follow-up of most datasets. EXPERIMENTAL DESIGN: We performed targeted panel sequencing covering 125 myeloma-specific genes and the loci involved in translocations in 223 newly diagnosed myeloma samples recruited into one of the total therapy trials. RESULTS: As expected, the most commonly mutated genes were NRAS, KRAS, and BRAF, making up 44% of patients. Double-Hit and BRAF and DIS3 mutations had an impact on outcome alongside classical risk factors in the context of an intensive treatment approach. We were able to identify both V600E and non-V600E BRAF mutations, 58% of which were predicted to be hypoactive or kinase dead. Interestingly, 44% of the hypoactive/kinase dead BRAF-mutated patients showed co-occurring alterations in KRAS, NRAS, or activating BRAF mutations, suggesting that they play a role in the oncogenesis of multiple myeloma by facilitating MAPK activation and may lead to chemoresistance. CONCLUSIONS: Overall, these data highlight the importance of mutational screening to better understand newly diagnosed multiple myeloma and may lead to patient-specific mutation-driven treatment approaches.


Asunto(s)
Biomarcadores de Tumor/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Mieloma Múltiple/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Análisis Mutacional de ADN , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Pronóstico , Tasa de Supervivencia
11.
Cell Biol Int ; 33(7): 765-71, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19422927

RESUMEN

We have investigated the effect of adiponectin (APN) peptide II on new vessel growth in mouse model of choroidal neovascularization (CNV) or wet type age-related macular degeneration (AMD). Mice were injected intraperitoneally with APN peptide II, control peptide, or PBS on day 1-7 or day 5-14. APN, AdipoR1, PCNA, and VEGF localization was investigated using confocal microscopy, immunohistochemistry, and RT-PCR. APN peptide II decreased the relative area of FITC-dextran perfused vessels by 4-fold, PCNA expression by 3-fold, and the number of PCNA stained HUVEC and MAVEC cells by 38 and 46%, respectively. We concluded that APN peptide II inhibits CNV size on days 7 and 14 by inhibiting the proliferation of endothelial cells in vivo and in vitro. APN peptide II may have therapeutic potential to inhibit CNV or wet AMD.


Asunto(s)
Adiponectina/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Adiponectina/química , Secuencia de Aminoácidos , Animales , Neovascularización Coroidal/etiología , Neovascularización Coroidal/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , Rayos Láser , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Fragmentos de Péptidos/química , Antígeno Nuclear de Célula en Proliferación/metabolismo , Antígeno Nuclear de Célula en Proliferación/fisiología , Estructura Terciaria de Proteína , Receptores de Adiponectina/metabolismo , Receptores de Adiponectina/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología
12.
Oncotarget ; 10(7): 732-737, 2019 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-30774775

RESUMEN

Hyperhaploid multiple myeloma is a rare numerical aberration group defined by a range of 24-34 chromosomes, which is associated with a poor prognosis with a 5-year survival rate of 23%. Hyperhaploid patient samples (n=8) were sequenced and copy number and mutations identified. Samples had a median of 13 monosomies (range 12-14), which in general were those not associated with trisomies in hyperdiploid samples. The chromosomes traditionally trisomic in hyperdiploid myeloma were disomic in hyperhaploid myeloma with retention of heterodisomy. We examined the hyperhaploid samples for frequently mutated genes and found that 8/8 (100%) hyperhaploid samples had a mutation in TP53, exceeding the overall rate of mutation in newly diagnosed patients (5.5%), indicating an oncogenic dependency in this group. All samples with TP53 mutation also had monosomy of chromosome 17, indicating bi-allelic inactivation of TP53. As such, this high risk group is part of double-hit myeloma.

13.
FEBS Lett ; 582(23-24): 3451-8, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18789935

RESUMEN

The objective of the present study was to investigate the effect of alcohol and nicotine consumption on the pathogenesis of choroidal neovascularization (CNV) in rats after laser-photocoagulation. Confocal microscopic analysis demonstrated an increase in CNV complex size in rats fed with alcohol (2.3-fold), nicotine (1.9-fold), and the combination of alcohol and nicotine (2.7-fold) compared with the control groups. Immunohistochemical analysis revealed that alcohol and nicotine consumption increased MAC deposition and VEGF expression in laser spots. Expression of CD59 by RT-PCR and Western blot was drastically reduced in the animals that were fed with alcohol, nicotine and alcohol and nicotine compared to those fed with water alone and this was associated with exacerbation of CNV.


Asunto(s)
Consumo de Bebidas Alcohólicas/patología , Neovascularización Coroidal/patología , Etanol/toxicidad , Nicotina/toxicidad , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Neovascularización Coroidal/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Masculino , Ratas , Ratas Endogámicas BN
14.
FEBS Lett ; 581(10): 1977-82, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17466298

RESUMEN

The aim of this study was to investigate the role of adiponectin (APN) in a mouse model of laser induced choroidal neovascularization (CNV). We have shown by immunohistochemistry that the expression of APN, adiponectin receptor 1, adiponectin receptor 2 and T cadherin gradually increased from day 1 to day 7 post-laser in laser treated mice compared to controls. Recombinant APN (rAPN) was injected intraperitoneally (i.p., 25 microg/mouse) or intravitreally (2 microg/eye) in lasered mice. Another set of lasered mice received APN peptide via i.p. (75 microg/mouse) or intravitreal (30 microg/eye) route. Control mice received a similar treatment with PBS, control protein or control peptide after laser treatment. We found that in the i.p. and intravitreal injection of rAPN resulted in 78% and 68% inhibition respectively in the size of CNV complex compared to control mice. Similar results were observed when APN peptide was injected intravitreally or i.p. Treatment with rAPN or the peptide resulted in decreased levels of vascular endothelial growth factor. Thus, APN inhibited choroidal angiogenesis and may have therapeutic implications in the treatment of wet age related macular degeneration.


Asunto(s)
Adiponectina/metabolismo , Coroides/irrigación sanguínea , Coroides/metabolismo , Neovascularización Coroidal/patología , Adiponectina/farmacología , Animales , Neovascularización Coroidal/inducido químicamente , Regulación de la Expresión Génica/efectos de los fármacos , Sustancias de Crecimiento/metabolismo , Rayos Láser , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Péptidos/química , Péptidos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA