Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Theriogenology ; 225: 119-129, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38805994

RESUMEN

Endometrosis in mares is a disease resulting from chronic inflammation characterized by peri glandular fibrosis. There is no effective treatment so far, which opens the door for exploring the use of stem cells as a candidate. Transforming growth factor beta (TGFß) is crucial for the establishment and progression of fibrosis in mare's endometrosis. We aimed to develop regenerative approaches to treat endometrosis by using mesenchymal stem cells (MSC), for which understanding the effect of TGFß on exogenous MSC is crucial. We isolated and characterized equine adipose MSC from six donors. Cells were pooled and exposed to 10 ng/ml of TGFß for 0, 4, and 24 h, after which cells were analyzed for proliferation, migration, mesodermal differentiation, expression of fibrosis-related mRNAs, and prostaglandin E2 secretion. At 24 h of exposition to TGFß, there was a progressive increase in the contraction of the monolayer, leading to nodular structures, while cell viability did not change. Exposure to TGFß impaired adipogenic and osteogenic differentiation after 4 h of treatment, which was more marked at 24 h, represented by a decrease in Oil red and Alizarin red staining, as well as a significant drop (p < 0.05) in the expression of key gene regulators of differentiation processes (PPARG for adipose and RUNX2 for osteogenic differentiation). TGFß increased chondrogenic differentiation as shown by the upsurge in size of the resulting 3D cell pellet and intensity of Alcian Blue staining, as well as the significant up-regulation of SOX9 expression (p < 0.05) at 4 h, which reached a maximum peak at 24 h (p < 0.01), indicative of up-regulation of glycosaminoglycan synthesis. Preconditioning MSC with TGFß led to a significant increase (p < 0.05) in the expression of myofibroblast gene markers aSMA, COL1A1, and TGFß at 24 h exposition time. In contrast, the expression of COL3A1 did not change with respect to the control but registered a significant downregulation compared to 4 h (p < 0.05). TGFß also affected the expression of genes involved in PGE2 synthesis and function; COX2, PTGES, and the PGE2 receptor EP4 were all significantly upregulated early at 4 h (p < 0.05). Cells exposed to TGFß showed a significant upregulation of PGE2 secretion at 4 h compared to untreated cells (p < 0.05); conversely, at 24 h, the PGE2 values decreased significantly compared to control cells (p < 0.05). Preconditioning MSC for 4 h led to an anti-fibrotic secretory phenotype, while a longer period (24 h) led to a pro-fibrotic one. It is tempting to propose a 4-h preconditioning of exogenous MSC with TGFß to drive them towards an anti-fibrotic phenotype for cellular and cell-free therapies in fibrotic diseases such as endometrosis of mares.


Asunto(s)
Tejido Adiposo , Enfermedades de los Caballos , Células Madre Mesenquimatosas , Factor de Crecimiento Transformador beta , Animales , Caballos , Femenino , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Enfermedades de los Caballos/terapia , Enfermedades de los Caballos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Diferenciación Celular/efectos de los fármacos , Fibrosis , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos
2.
Vet Q ; 44(1): 1-11, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39086189

RESUMEN

Mare endometrosis is a major reproductive problem associated with low fertility and is characterized by persistent inflammation, TGFß-1 signaling, and consequently, extracellular matrix deposition, which compromises endometrial glands. Mesenchymal stem cell-based products (MSCs), such as extracellular vesicles (EVs), have gained attention due to the regulatory effects exerted by their miRNA cargo. Here, we evaluated the impact of preconditioning equine adipose mesenchymal stem cells with TGFß-1 for short or long periods on the anti-fibrotic properties of secreted extracellular vesicles. MSCs were isolated from six healthy horses and exposed to TGFß-1 for 4, 24, and 0 h. The expression of anti-fibrotic and pro-fibrotic miRNAs and mRNAs in treated cells and miRNAs in the cargo of secreted extracellular vesicles was measured. The resulting EVs were added for 48 h to endometrial stromal cells previously induced to a fibrotic status. The expression of anti-fibrotic and pro-fibrotic genes and miRNAs was evaluated in said cells using qPCR and next-generation sequencing. Preconditioning MSCs with TGFß-1 for 4 h enriched the anti-fibrotic miRNAs (mir29c, mir145, and mir200) in cells and EVs. Conversely, preconditioning the cells for 24 h leads to a pro-fibrotic phenotype overexpressing mir192 and mir433. This finding might have implications for developing an EV-based protocol to treat endometrial fibrosis in mares.


Asunto(s)
Endometrio , Vesículas Extracelulares , Fibrosis , Células Madre Mesenquimatosas , MicroARNs , Factor de Crecimiento Transformador beta1 , Animales , Caballos , Femenino , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Vesículas Extracelulares/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Endometrio/metabolismo , Endometrio/citología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Células del Estroma/metabolismo , Células del Estroma/efectos de los fármacos , Enfermedades de los Caballos , Regulación de la Expresión Génica/efectos de los fármacos , Endometriosis/veterinaria , Endometriosis/metabolismo , Endometriosis/genética
3.
Front Vet Sci ; 10: 1271240, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37869492

RESUMEN

The modulation of inflammation is pivotal for uterine homeostasis. Here we evaluated the effect of the oestrus cycle on the expression of pro-inflammatory and anti-inflammatory markers in a cellular model of induced fibrosis. Mare endometrial stromal cells isolated from follicular or mid-luteal phase were primed with 10 ng/mL of TGFß alone or in combination with either IL1ß, IL6, or TNFα (10 ng/mL each) or all together for 24 h. Control cells were not primed. Messenger and miRNA expression were analyzed using real-time quantitative PCR (RT-qPCR). Cells in the follicular phase primed with pro-inflammatory cytokines showed higher expression of collagen-related genes (CTGF, COL1A1, COL3A1, and TIMP1) and mesenchymal marker (SLUG, VIM, CDH2, and CDH11) genes; p < 0.05. Cells primed during the mid-luteal overexpressed genes associated with extracellular matrix, processing, and prostaglandin E synthase (MMP2, MMP9, PGR, TIMP2, and PTGES; p < 0.05). There was a notable upregulation of pro-fibrotic miRNAs (miR17, miR21, and miR433) in the follicular phase when the cells were exposed to TGFß + IL1ß, TGFß + IL6 or TGFß + IL1ß + IL6 + TNFα. Conversely, in cells from the mid-luteal phase, the treatments either did not or diminished the expression of the same miRNAs. On the contrary, the anti-fibrotic miRNAs (miR26a, miR29b, miR29c, miR145, miR378, and mir488) were not upregulated with treatments in the follicular phase. Rather, they were overexpressed in cells from the mid-luteal phase, with the highest regulation observed in TGFß + IL1ß + IL6 + TNFα treatment groups. These miRNAs were also analyzed in the extracellular vesicles secreted by the cells. A similar trend as seen with cellular miRNAs was noted, where anti-fibrotic miRNAs were downregulated in the follicular phase, while notably elevated pro-fibrotic miRNAs were observed in extracellular vesicles originating from the follicular phase. Pro-inflammatory cytokines may amplify the TGFß signal in the follicular phase resulting in significant upregulation of extracellular matrix-related genes, an imbalance in the metalloproteinases, downregulation of estrogen receptors, and upregulation of pro-fibrotic factors. Conversely, in the luteal phase, there is a protective role mediated primarily through an increase in anti-fibrotic miRNAs, a decrease in SMAD2 phosphorylation, and reduced expression of fibrosis-related genes.

4.
Medicina (B Aires) ; 66(5): 421-6, 2006.
Artículo en Español | MEDLINE | ID: mdl-17137171

RESUMEN

In patients with porphyria cutanea tarda (PCT), hepatic iron accumulation associated to hereditary hemochromatosis (HH) could play a role in the etiology and in the clinical expression of the disease. The H63D and C282Y mutations of the HFE gene frequency were studied in a PCT group of patients and compared with the frequency observed in a group of volunteer blood donors. PCT patients were cataloged as hereditary or acquired PCT carriers, whether or not they presented uroporphyrinogen decarboxilase gene mutations. Fifty percent of PCT patients were carriers of the disease's genetic type. Such percentage is significantly higher than what other authors have previously informed. H63D and C282Y mutations were present in 23% and 2.4% of the volunteer blood donors, respectively. Similar frequencies were informed by others authors in Chilean white ethnic populations, and also in Spaniard and Argentinean populations, but significantly higher than that observed in Chile's Araucanean aboriginal population. Probably the frequency of H63D and C283Y mutations are related to the Spaniard ascendancy dominance of Chile's white ethnic population. The frequency of HFE gene mutations in PCT patients was not different than what was observed in volunteer blood donors. Similarly, there was no statistical difference in the frequency of these mutations among patients with acquired or genetic PCT disease. With the obtained results, it is not possible postulate an association between PCT and the hereditary hemochromatosis of HFE gene mutations carrier conditions.


Asunto(s)
Donantes de Sangre , Hemocromatosis/genética , Mutación , Porfiria Cutánea Tardía/genética , Chile/etnología , Femenino , Frecuencia de los Genes , Tamización de Portadores Genéticos , Genotipo , Hemocromatosis/sangre , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Sobrecarga de Hierro , Masculino , Proteínas de la Membrana/genética , Porfiria Cutánea Tardía/sangre , Uroporfirinógeno Descarboxilasa/sangre , Uroporfirinógeno Descarboxilasa/genética
5.
Parasitol. día ; 15(3/4): 117-21, jul.-dic. 1991. tab
Artículo en Español | LILACS | ID: lil-109462

RESUMEN

Se realizó un estudio clínico serológico prospectivo en 200 niños con patología oncológica para determinar la frecuencia y riesgo de toxoplasmosis en esta población. El estudio se efectuó por reacción de inmunofluorescencia indirecta para T. gondii, considerándose títulos positivos aquellos * 1:16. De ellos, 75 (37,5%) presentaron serología positiva, 53 (26,5%) títulos bajos, 15 (7,5%) títulos medianos y 7 (3,5%) títulos altos. De acuerdo a la patología de base 41,6% de los tumores óseos presentó toxoplasmosis, el 35,2% de las leucemias y el 30% de los linfomas. Todos los niños seropositivos fueron controlados periódicamente mediante examen clínico y relación de inmunofluorescencia indirecta en suero. Se demuestra que los niños oncológicos tienen un riesgo 2,86 veces mayor de presentar toxoplasmosis (p < 0,05) con un intervalo de confianza de 1,884,32; aquéllos con tumores óseos presentan un riesgo 4,4 veces mayor. En relación a lo descrito para población normal, en cambio, los pacientes portadores de linfoma no presentaron mayor riesgo de toxoplasmosis que la población pediátrica sin esta patología


Asunto(s)
Lactante , Preescolar , Niño , Adolescente , Humanos , Masculino , Femenino , Neoplasias/parasitología , Toxoplasmosis/epidemiología , Toxoplasmosis/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA