Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Theriogenology ; 225: 119-129, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38805994

RESUMO

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.


Assuntos
Tecido Adiposo , Doenças dos Cavalos , Células-Tronco Mesenquimais , Fator de Crescimento Transformador beta , Animais , Cavalos , Feminino , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Doenças dos Cavalos/terapia , Doenças dos Cavalos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia , Diferenciação Celular/efeitos dos fármacos , Fibrose , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos
2.
Cell Reprogram ; 22(6): 311-327, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32991224

RESUMO

Equine endometrial and adipose mesenchymal stem cells (eMSCs and aMSCs, respectively) were isolated from the same donors of thoroughbred mares. The cells displayed characteristic features of MSCs, including trilineage mesodermal and also neurogenic differentiation. We evaluated the influence of cellular origin on their transcriptome profile. Cellular RNA was isolated and sequenced and extracellular vesicles (EVs) were obtained from conditioned medium of cells cultured in medium depleted of EVs, and their microRNA (miRNA) cargo analyzed by sequencing. Differential expression of mRNAs and EV-miRNA was analyzed, as well as pathways and processes most represented in each cell origin. mRNA reads from all expressed genes clustered according to the cellular origin. A total of 125 up- and 51 downregulated genes were identified and 31 differentially expressed miRNAs. Based on mRNA sequencing, endometrial MSCs strongly upregulated genes involved in the Hippo, transforming growth factor beta, and pluripotency signaling pathways. Alongside with this, pathways involved in extracellular matrix reorganization were the most represented in the miRNA cargo of EVs secreted by eMSCs. The niche from which MSCs originated defined the transcriptomic signature of the cells, including the secretion of lineage-specific loaded EV to ensure proper communication and homeostasis. Identification and testing their biological functions can provide new tools for the therapeutic use of horse MSC.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular , Endométrio/citologia , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Transcriptoma , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Endométrio/metabolismo , Vesículas Extracelulares/genética , Feminino , Cavalos , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais
3.
Theriogenology ; 155: 33-42, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32622203

RESUMO

Horse mesenchymal stem cells (MSC) are potential anti-inflammatory tools for post-breeding induced endometritis (PBIE). In this research MSCs isolated from the endometrium or subcutaneous fat of the same donors were infused iu into mares with PBIE for assessment of their anti-inflammatory action and engraftment. PBIE was induced in nine gynecologically healthy mares by iu infusion of 500 million dead sperm in saline. Inflammatory markers were analyzed in uterine lavages and biopsies immediately before (phase I) and 3 h after infusion of sperm (phase II). Measurements: polymorph nuclear cells (PMN), proteins IL-6 and TNFα (ELISA in the lavages) and immunostaining in biopsies, transcripts of IL-1α, 6, 8, 10, TNFα and COX2 (qPCR of pelleted lavages). At 24 h after sperm deposition (phase III), mares were instilled iu with 20 ml of saline containing 2 × 107 adipose MSCs (n = 3, group 1) or endometrial MSCs (n = 3, group 2). Cells were labeled previously with carboxyfluorescein diacetate succinimidyl ester (CFDA SE). A third group (n = 3) received 20 mL of sterile saline alone. After 48 h another biopsy/lavage were done and the same parameters analyzed. For engraftment, additional biopsies were taken at days 10 and 30 of sperm infusion and analyzed by confocal microscopy. Dead sperm in saline markedly increased PMNs counts, IL-6 and TNFα expression in the ELISA (p < 0.05) and immunostaining. In phase III a significant reduction (p < 0.0001) of PMN was found in all samples, including control mares. A decrease (p < 0.05) of IL-6 and TNF-α was detected by ELISA, in the groups that received MSC, but not in control group. In the aMSC-treated group, a significant decrease was found in the expression of (IL1α, p = 0.0003; IL-6 p 0.04; IL-8, p = 0.006, TNFα p = 0.004). Expression of IL-10 and COX2 remained unchanged (p = 0.08). In the mares that received eMSC, IL-6 and 8 decreased significantly (p = 0.01), IL-10 increased (p = 0.009), while TNFα, COX2 and IL1α did not significantly change their expression. In the engraftment experiment CFDA label was found sparingly in all the samples analyzed until day 30, mainly at the stromal compartment of the endometrium. No differences in the engraftment pattern was found among cell origins. We conclude that inoculation of MSCs significantly reduced inflammation independently of the origin of the cells and that cells perform limited engraftment detectable after one month of infusion. These findings can be of help for the design of new anti-inflammatory therapies of uterine diseases in mares.


Assuntos
Endometrite , Doenças dos Cavalos , Células-Tronco Mesenquimais , Animais , Anti-Inflamatórios , Endometrite/tratamento farmacológico , Endometrite/veterinária , Endométrio , Feminino , Doenças dos Cavalos/tratamento farmacológico , Cavalos
4.
Animals (Basel) ; 10(6)2020 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-32585798

RESUMO

In mesenchymal stem cells (MSCs), it has been reported that prostaglandin E2 (PGE2) stimulation of EP2 and EP4 receptors triggers processes such as migration, self-renewal, survival, and proliferation, and their activation is involved in homing. The aim of this work was to establish a genetically modified adipose (aMSC) model in which receptor genes EP2 and EP4 were edited separately using the CRISPR/Cas9 system. After edition, the genes were evaluated as to if the expression of MSC surface markers was affected, as well as the migration capacity in vitro of the generated cells. Adipose MSCs were obtained from Chilean breed horses and cultured in DMEM High Glucose with 10% fetal bovine serum (FBS). sgRNA were cloned into a linearized LentiCRISPRv2GFP vector and transfected into HEK293FT cells for producing viral particles that were used to transduce aMSCs. GFP-expressing cells were separated by sorting to obtain individual clones. Genomic DNA was amplified, and the site-directed mutation frequency was assessed by T7E1, followed by Sanger sequencing. We selected 11 clones of EP2 and 10 clones of EP4, and by Sanger sequencing we confirmed 1 clone knock-out to aMSC/EP2 and one heterozygous mutant clone of aMSC/EP4. Both edited cells had decreased expression of EP2 and EP4 receptors when compared to the wild type, and the edition of EP2 and EP4 did not affect the expression of MSC surface markers, showing the same pattern in filling the scratch. We can conclude that the edition of these receptors in aMSCs does not affect their surface marker phenotype and migration ability when compared to wild-type cells.

5.
Stem Cell Res Ther ; 10(1): 135, 2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31109365

RESUMO

BACKGROUND: Xenotransplantation of spermatogonial stem cells (SSCs) has become a popular topic in various research fields because manipulating these cells can provide insights into the mechanisms associated with germ cell lines and the entire spermatogenesis process; moreover, these cells can be used in several biotechnology applications. To achieve successful xenotransplantation, the in vitro microenvironment in which SSCs are cultured should be an ideal microenvironment for self-renewal and similar to the in vivo testicular microenvironment. The age of the donor, the correct spermatogenesis cycle, and the quality of the donor tissue are also important. Although cell culture-related factors, such as the in vitro supplementation of hormonal factors, are known to promote successful xenotransplantation in mice, little is known about the influence of these factors on SSCs in vitro or in vivo in other mammalian species, such as dogs (Canis lupus familiaris). In this context, the goals of this study were to test the effect of follicle-stimulating hormone (FSH) on canine spermatogonial stem cell (cSSC) cultures since this hormone is related to the glial cell-derived neurotrophic factor (GDNF) signaling pathway, which is responsible for the self-renewal and maintenance of these cells in vivo, and to investigate the microenvironment of the SSC culture after FSH supplementation. Additionally, in vivo analyses of transplanted FSH-supplemented cSSCs in the testes of infertile mice were performed to assess the capacity of cSSCs to develop, maintain, and restore spermatogenesis. METHODS: SSCs from canine prepubertal testes (aged 3 months) were cultured in vitro in the presence of FSH (10 IU L-1). GFRA1 transcript expression was detected to confirm the spermatogonia population in culture and the effect of FSH on these cells. The protein and transcript levels of late germ cell markers (GFRA1, DAZL, STRA8, PLZF, and CD49f) and a pluripotency marker (OCT4) were detected at 72 and 120 h to confirm the cSSC phenotype. In vivo experiments were performed by transplanting GFP+ cSSCs into infertile mice, and a 10-week follow-up was performed. Histological and immunofluorescence analyses were performed to confirm the repopulation capacity after cSSC xenotransplantation in the testis. RESULTS: Supplementation with FSH in cell culture increased the number of cSSCs positive for GFRA1. The cSSCs were also positive for the pluripotency and early germline marker OCT4 and the late germline markers PLZF, DAZL, C-kit, and GFRA-1. The in vivo experiments showed that the cSSCs xenotransplanted into infertile mouse testes were able to repopulate germline cells in the seminiferous tubules of mice. CONCLUSIONS: In conclusion, our results showed for the first time that the treatment of cSSC cultures with FSH can promote in vitro self-renewal, increase the population of germline cells, and possibly influence the success of spermatogenesis in infertile mice in vivo.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Espermatogênese/genética , Espermatogônias/transplante , Transplante Heterólogo/métodos , Animais , Cães , Masculino , Camundongos , Espermatogônias/citologia
6.
Pesqui. vet. bras ; 31(6): 538-542, jun. 2011. ilus
Artigo em Português | LILACS, Sec. Est. Saúde SP | ID: lil-593234

RESUMO

The aim of the present study was to evaluate the cell proliferative activity, by AgNORs number, in different regions of bovine placenta throughout gestation. A total of 28 bovine placentas were separated into four groups: group I (60 to 120 days), group II (121 to 170 days), group III (171 to 220 days), and group IV (221 to 290 days). It was found a greater number of AgNORs in giant trophoblastic cells (GTC) when compared with mononuclear trophoblastic cells (MTC) (p<0,001) in all regions and gestational groups analyzed, that confirms their intensive synthesis activity in trophoblast epithelium. The central region of the placentome begins an intense proliferative activity in group II, observed by clusters, while placentomes edges showed a higher number of clusters on group III. These data suggest that the central region of the placentomes began an intense proliferative activity prior to its edge, both declines at the end of pregnancy. Interplacentomal area showed a higher number of AgNORs in the group IV, suggesting a higher proliferative activity of these cells at the end of pregnancy. The results of this study indicate that the proliferative activity, as determined by the amount of intranuclear AgNORs, exhibits patterns that are not only specific to each type of trophoblastic cells, but also for each specific region of bovine placenta throughout pregnancy.


Este estudo teve como objetivo analisar atividade proliferativa das células trofoblásticas, através da quantificação de AgNORs, em diferentes regiões da placenta bovina ao longo da gestação. Foram utilizados 28 úteros, sendo estes agrupados de acordo com as idades gestacionais: grupo I (60-120 dias); II (121- 170 dias); III (171-220 dias) e IV (221-290 dias). Foi encontrado um número significativamente maior de AgNORs nas células trofoblásticas gigantes (CTG) em relação às mononucleadas (CTM) (p<0,001) em todas as regiões e grupos gestacionais analisados, o que confirma sua intensa atividade de síntese no epitélio trofoblástico. A região central do placentônio inicia uma atividade proliferativa mais intensa já no grupo II, observada pelo número de clusters, enquanto que a margem do placentônio apresenta uma maior quantidade de clusters no grupo III. Estes dados sugerem que a região central do placentônio inicia uma intensa atividade proliferativa anteriormente a sua margem, ambas declinando no final da gestação. A área interplacentomal apresentou um maior número de AgNORs no último grupo gestacional, sugerindo uma maior atividade proliferativa dessas células no final da prenhez. Os resultados deste estudo indicam que a atividade proliferativa, determinada pela quantidade de AgNORs intranucleares, exibe padrões que são específicos não somente para cada tipo de célula trofoblástica, mas também para cada região específica da placenta bovina ao longo da gestação.


Assuntos
Animais , Gravidez , Bovinos , Doença Trofoblástica Gestacional , Tumor Trofoblástico de Localização Placentária/diagnóstico , Tumor Trofoblástico de Localização Placentária/veterinária , Proliferação de Células
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...