Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119806, 2024 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-39098401

RESUMEN

Nowadays, regenerative medicine techniques are usually based on the application of mesenchymal stromal cells (MSCs) for the repair or restoration of injured damaged tissues. However, the effectiveness of autologous therapy is limited as therapeutic potential of MSCs declines due to patient's age, health condition and prolonged in vitro cultivation as a result of decreased growth rate. For that reason, there is an urgent need to develop strategies enabling the in vitro rejuvenation of MSCs prior transplantation in order to enhance their in vivo therapeutic efficiency. In presented study, we attempted to mimic the naturally occurring mitochondrial transfer (MT) between neighbouring cells and verify whether artificial MT (AMT) could reverse MSCs aging and improve their biological properties. For that reason, mitochondria were isolated from healthy donor equine adipose-derived stromal cells (ASCs) and transferred into metabolically impaired recipient ASCs derived from equine metabolic syndrome (EMS) affected horses, which were subsequently subjected to various analytical methods in order to verify the cellular and molecular outcomes of the applied AMT. Mitochondria recipient cells were characterized by decreased apoptosis, senescence and endoplasmic reticulum stress while insulin sensitivity was enhanced. Furthermore, we observed increased mitochondrial fragmentation and associated PARKIN protein accumulation, which indicates on the elimination of dysfunctional organelles via mitophagy. AMT further promoted physioxia and regulated autophagy fluxes. Additionally, rejuvenated ASCs displayed an improved anti-inflammatory activity toward LPS-stimulated synoviocytes. The presented findings highlight AMT as a promising alternative and effective method for MSCs rejuvenation, for potential application in autologous therapies in which MSCs properties are being strongly deteriorated due to patients' condition.

2.
J Mol Med (Berl) ; 102(8): 1015-1036, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38874666

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) is a transcription factor that promotes adipogenesis, lipid uptake and storage, insulin sensitivity, and glucose metabolism. Hence, defects in PPARγ have been associated to the development of metabolic disorders. Sex hormone-binding globulin (SHBG) is a glycoprotein primarily produced in the liver that regulates the bioavailability of sex hormones. Alike PPARγ, low SHBG levels have been correlated with insulin resistance and associated endocrine abnormalities. Therefore, this study aimed to verify whether SHBG may restore depleted PPARγ functions and thus serve as a new candidate for the management of metabolic conditions. A model of equine adipose-derived stromal cells (EqASCs) has been used, in which a PPARγ silencing and SHBG treatment have been achieved to determine the changes in cell viability, premature senescence, oxidative stress, and mitochondrial functions. Obtained data demonstrated that loss in PPARγ triggers cell apoptosis which is not reversed by SHBG application. Moreover, PPARγ knockdown cells exhibited premature senescence, which has been substantially alleviated by SHBG concomitantly to increased BAX/BCL2 ratio, suggesting a possible effect on senescence-induced apoptosis resistance. Interestingly, PPARγ silencing induced a significant alteration in mitochondrial membrane potential as well as the expression of dynamics and metabolism-related markers. SHBG treatment enabled to ameliorate the transmembrane potential, to normalize the expression levels of key dynamics and metabolism mediators, and to restore the protein levels of PINK, which is critically involved in mitochondria recycling machinery. Presented data suggest that SHBG may provide new mechanistic insights into the regulation of PPARγ functions, and thus offers a preliminary picture on a possible SHBG-PPARγ metabolic crosstalk. KEY MESSAGES : PPARγ is a transcription factor that tightly regulates cell metabolism. Low SHBG levels correlate with insulin resistance and associated endocrine abnormalities. PPARγ silencing reduces cell viability, triggers premature senescence and profound mitochondrial failure in equine ASCs. SHBG protein reverses senescent phenotype and apoptosis resistance of PPARγ- ASCs. SHBG improves mitochondrial dynamics and metabolism following PPARγ knockdown. SHBG might serve as a PPARγ potential mimicking agent for the modulation of ASCs metabolic processes.


Asunto(s)
Apoptosis , Dinámicas Mitocondriales , PPAR gamma , Globulina de Unión a Hormona Sexual , Células del Estroma , Animales , PPAR gamma/metabolismo , Caballos , Globulina de Unión a Hormona Sexual/metabolismo , Globulina de Unión a Hormona Sexual/genética , Células del Estroma/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Tejido Adiposo/metabolismo , Tejido Adiposo/citología , Supervivencia Celular , Senescencia Celular , Potencial de la Membrana Mitocondrial , Células Cultivadas
3.
Front Endocrinol (Lausanne) ; 15: 1382844, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38689728

RESUMEN

Equine metabolic syndrome (EMS) is a critical endocrine condition in horses, characterized by hyperinsulinemia, hyperlipidemia, and insulin resistance, posing a significant threat to their health. This study investigates the efficacy of supplementing EMS-affected horses with Arthrospira platensis enriched with Cr(III), Mg(II), and Mn(II) ions using biosorption process in improving insulin sensitivity and glucose tolerance, reducing inflammation, and mitigating obesity-related fat accumulation. Our results demonstrate that Arthrospira supplementation reduces baseline insulin and glucose levels, contributing to decreased adipose tissue inflammation. Furthermore, Arthrospira supplementation results in a decrease in body weight and improvements in overall body condition scores and cresty neck scores. Additionally, administration of Arthrospira leads to reduced levels of triglycerides and aspartate aminotransferase, indicating a decrease in hepatic adiposity and inflammation. These findings suggest that Arthrospira, enriched with essential micro- and macroelements, can be an advanced feed additive to enhance insulin sensitivity, promote weight reduction, and alleviate inflammatory processes, thereby improving the overall condition of horses affected by EMS. The use of Arthrospira as a feed additive has the potential to complement conventional management strategies for EMS.


Asunto(s)
Alimentación Animal , Cromo , Suplementos Dietéticos , Enfermedades de los Caballos , Inflamación , Resistencia a la Insulina , Magnesio , Manganeso , Síndrome Metabólico , Spirulina , Animales , Caballos , Inflamación/metabolismo , Síndrome Metabólico/veterinaria , Síndrome Metabólico/metabolismo , Enfermedades de los Caballos/metabolismo , Enfermedades de los Caballos/prevención & control , Alimentación Animal/análisis , Magnesio/metabolismo , Masculino , Femenino
4.
Curr Issues Mol Biol ; 46(4): 3251-3277, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38666934

RESUMEN

Corneal ulcers, characterized by severe inflammation of the cornea, can lead to serious, debilitating complications and may be vision-threatening for horses. In this study, we aimed to investigate the role of endoplasmic reticulum (ER) stress in corneal stem progenitor cell (CSSC) dysfunction and explore the potential of equine adipose-derived stromal stem cell (ASC)-derived extracellular vesicles (EVs) to improve corneal wound healing. We showed that CSSCs expressed high levels of CD44, CD45, and CD90 surface markers, indicating their stemness. Supplementation of the ER-stress-inducer tunicamycin to CSSCs resulted in reduced proliferative and migratory potential, accumulation of endoplasmic reticulum (ER)-stressed cells in the G0/G1 phase of the cell cycle, increased expression of proinflammatory genes, induced oxidative stress and sustained ER stress, and unfolded protein response (UPR). Importantly, treatment with EVs increased the proliferative activity and number of cells in the G2/Mitosis phase, enhanced migratory ability, suppressed the overexpression of proinflammatory cytokines, and upregulated the anti-inflammatory miRNA-146a-5p, compared to control and/or ER-stressed cells. Additionally, EVs lowered the expression of ER-stress master regulators and effectors (PERK, IRE1, ATF6, and XBP1), increased the number of mitochondria, and reduced the expression of Fis-1 and Parkin, thereby promoting metabolic homeostasis and protecting against apoptosis in equine CSSCs. Our findings demonstrate that MSCs-derived EVs represent an innovative and promising therapeutic strategy for the transfer of bioactive mediators which regulate various cellular and molecular signaling pathways.

5.
J Am Vet Med Assoc ; 262(S1): S31-S39, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38479108

RESUMEN

OBJECTIVE: Extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) are promising avenues in regenerative medicine, offering unique immunomodulatory and regenerative properties with lower immunogenicity. This study delves into the distinctive features of EVs extracted from feline adipose-derived MSCs (ASCs) and placenta-derived MSCs (PMSCs). The tissues were collected from 11 female cats aged between 4 and 7 years old. SAMPLE: EVs extracted from MSCs from discarded fetal membranes from 7 female cats and SC adipose tissue from 11 cats. METHODS: We comprehensively explored morphological characteristics, mitochondrial density, surface markers, and pro- and anti-inflammatory mediators, uncovering notable differences between ASCs and PMSCs. RESULTS: Morphologically, ASCs exhibit a spindle-shaped form in contrast to the spherical morphology of PMSCs. Proliferation and clonogenic potential assessments reveal the faster proliferation and robust clonogenic nature of ASCs, suggesting their potential vital role in regenerative processes. Surface marker expression analysis indicates a significantly higher expression of multipotency-associated markers in ASCs, suggesting their superior proregenerative potential. Phenotyping of EVs demonstrates distinctive features, with CD9 expression suggesting varied EV secretion patterns. Notably, PMSCs exhibit superior CD81 expression, indicating their potential as preferred donors of mitochondria. Pro- and anti-inflammatory mediators analyzed at mRNA and microRNA levels reveal higher RNA content in EVs compared to source cells, emphasizing the potential of EVs in directing regenerative processes. Differential microRNA expression in EVs derived from ASCs hints at their regulatory roles in anti-inflammatory and immunometabolic processes. CLINICAL RELEVANCE: This study lays a foundation for understanding the nuances between ASCs and PMSCs, which is crucial for harnessing the full therapeutic potential of MSCs and their EVs in tissue repair and regeneration.


Asunto(s)
Tejido Adiposo , Vesículas Extracelulares , Células Madre Mesenquimatosas , Placenta , Medicina Regenerativa , Animales , Femenino , Gatos , Vesículas Extracelulares/metabolismo , Placenta/citología , Tejido Adiposo/citología , Embarazo , Células Madre Mesenquimatosas/citología
6.
Cells ; 13(2)2024 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-38247843

RESUMEN

Equine metabolic syndrome (EMS) is a significant global health concern in veterinary medicine. There is increasing interest in utilizing molecular agents to modulate hepatocyte function for potential clinical applications. Recent studies have shown promising results in inhibiting protein tyrosine phosphatase (PTP1B) to maintain cell function in various models. In this study, we investigated the effects of the inhibitor Trodusquemine (MSI-1436) on equine hepatic progenitor cells (HPCs) under lipotoxic conditions. We examined proliferative activity, glucose uptake, and mitochondrial morphogenesis. Our study found that MSI-1436 promotes HPC entry into the cell cycle and protects them from palmitate-induced apoptosis by regulating mitochondrial dynamics and biogenesis. MSI-1436 also increases glucose uptake and protects HPCs from palmitate-induced stress by reorganizing the cells' morphological architecture. Furthermore, our findings suggest that MSI-1436 enhances 2-NBDG uptake by increasing the expression of SIRT1, which is associated with liver insulin sensitivity. It also promotes mitochondrial dynamics by modulating mitochondria quantity and morphotype as well as increasing the expression of PINK1, MFN1, and MFN2. Our study provides evidence that MSI-1436 has a positive impact on equine hepatic progenitor cells, indicating its potential therapeutic value in treating EMS and insulin dysregulation.


Asunto(s)
Colestanos , Resistencia a la Insulina , Síndrome Metabólico , Dinámicas Mitocondriales , Espermina , Animales , Glucosa , Caballos , Insulina/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Palmitatos , Espermina/análogos & derivados , Resistencia a la Insulina/fisiología
7.
Front Mol Biosci ; 10: 1214961, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38146533

RESUMEN

Equine metabolic syndrome (EMS) is a steadily growing endocrine disorder representing a real challenge in veterinary practice. As a multifactorial condition, EMS is characterized by three main metabolic abnormalities including insulin resistance, increased adiposity or obesity and hoof laminitis. Adipose tissue dysfunction is recognized as a core pathophysiological determinant of EMS, as it strongly participates to lipotoxicity and systemic metaflammation, both of which have been closely linked to the development of generalized insulin resistance. Besides, sex hormone binding globulin (SHBG) is an important sex steroids transporters that has been recently proposed as an important metabolic mediator. Therefore, the aim of this study was to verify whether SHBG treatment may ameliorate subcutaneous adipose tissue metabolic failure under EMS condition in terms of lipidome homeostasis, lipid metabolism programs, insulin signalling and local inflammation. Subcutaneous adipose tissue (SAT) biopsies were collected post-mortem from healthy (n = 3) and EMS (n = 3) slaughtered horses. SHBG protein has been applied to SAT samples from EMS horses for 24 h at a final concentration of 50 nM, while control groups (healthy and untreated EMS) were cultured in the presence of SHBG-vehicle only. Tissues from all groups were afterwards secured for downstream analysis of gene expression using RT-qPCR, protein levels by Western blot and ELISA assay and lipidomics through GC-MS technique. Obtained results showcased that SHBG intervention efficiently normalized the altered fatty acids (FAs) profiles by lowering the accumulation of saturated and trans FAs, as well as the pro-inflammatory arachidonic and linoleic acids. Moreover, SHBG showed promising value for the regulation of adipocyte lipolysis and engorgement by lowering the levels of perilipin-1. SHBG exerted moderated effect toward SCD1 and FASN enzymes expression, but increased the LPL abundance. Interestingly, SHBG exhibited a negative regulatory effect on pro-adipogenic stimulators and induced higher expression of KLF3, IRF3 and ß-catenin, known as strong adipogenesis repressors. Finally, SHBG protein showed remarkable ability in restoring the insulin signal transduction, IR/IRS/Pi3K/AKT phosphorylation events and GLUT4 transporter abundance, and further attenuate pro-inflammatory response by lowering IL-6 tissue levels and targeting the PDIA3/ERK axis. Overall, the obtained data clearly demonstrate the benefice of SHBG treatment in the regulation of adipose tissue metabolism in the course of EMS and provide new insights for the development of molecular therapies with potential translational application to human metabolic disorders.

8.
Int. j. morphol ; 31(3): 826-831, set. 2013. ilus
Artículo en Inglés | LILACS | ID: lil-694963

RESUMEN

Central nervous system of reptiles has the ability to grow and regenerate during adult life of the animal. Therefore, cells creating CNS of this animal class should compound substances or molecules enabling neuroregeneration. Cells directly involved in this process have not been clearly characterized, especially in cell culture environment. Morphology of reptilian glial adherent cells should be known better to find any differences from mammalian CNS cells. We isolated glial cells from olfactory bulb and cerebrum from gecko (Eublepharis macularius) and cultured separately. We have observed populations of cells with proliferative capacity in both types of cultures. Also, we have detected lipid molecules deposits within their cytoplasm, which localization was correlated with mitochondria position. This information can be helpful in searching new bioactive substances involved in regeneration of central nervous system.


El sistema nervioso central de los reptiles tiene la capacidad de crecer y regenerarse durante la vida adulta del animal. Por lo tanto, las células de SNC creadas de esta clase de animales deberían componerse de sustancias o moléculas que permiten la neuroregeneración. Las células que participan directamente en este proceso no han sido claramente caracterizadas, especialmente en el entorno de cultivo celular. La morfología de las células adherentes gliales de reptiles deben ser reconocidas y diferenciarse respecto a las células del SNC de mamíferos. Se aislaron células gliales del bulbo olfatorio y el cerebro del Gecko (Eublepharis macularius) y se cultivaron por separado. Se observaron poblaciones de células con capacidad proliferativa en ambos tipos de cultivos. Además, se detectaron moléculas de depósitos lipídicos dentro de su citoplasma, y su localización se correlacionó con la posición de las mitocondrias. Esta información puede ser útil en la búsqueda de nuevas sustancias bioactivas que participan en la regeneración del sistema nervioso central.


Asunto(s)
Animales , Lagartos/anatomía & histología , Neuroglía/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/crecimiento & desarrollo , Técnicas de Cultivo de Célula , Neurogénesis
9.
Int. j. morphol ; 29(3): 1012-1017, Sept. 2011. ilus
Artículo en Inglés | LILACS | ID: lil-608698

RESUMEN

Recent studies revealed multipotent properties of fat tissue isolated mesenchymal stem cells. These cells are successfully used as therapeutic factor for many locomotive disorders, being even more effective than stem cells from bone marrow. Isolated and cultured, AD-MSCs were observed, photographed and measured to compare cells from two different species.


Estudios recientes han revelado propiedades pluripotentes del tejido graso aislado de células madre mesenquimales. Estas células se utilizan con éxito como factor terapéutico para muchos trastornos locomotores, siendo aún más eficaz que las células madre de médula ósea. Aisladas y cultivadas, AD-MSC se observaron, fotografiaron y midieron comparar células de dos especies diferentes.


Asunto(s)
Perros , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/ultraestructura , Enfermedades Musculoesqueléticas/terapia , Enfermedades Musculoesqueléticas/veterinaria , Procedimientos Ortopédicos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...