Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Reprod Biomed Online ; 38(4): 579-587, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30777668

RESUMEN

RESEARCH QUESTION: Telomeres are non-coding, repetitive DNA sequences (TTAGGG repeats) that play an important role in maintaining genome integrity. Unlike in somatic cells, telomere length in spermatozoa increases with male age and is considered as a molecular marker of sperm quality. The aetiology of failed fertilization following intracytoplasmic sperm injection (ICSI) is multifactorial; perhaps one of the reasons for this failure in these individuals is shortened sperm telomere length. This study therefore aimed to assess sperm telomere length in addition to DNA damage, lipid peroxidation and protamine deficiency in infertile men with previously failed/low fertilization post-ICSI. DESIGN: Semen samples were obtained from infertile men with previous failed/low fertilization rates (n = 10). Chromatin integrity (chromomycin A3 staining and TUNEL assay), lipid peroxidation (BODIPY probe) and telomere length (real-time PCR) for semen samples from these men were compared with samples obtained from fertile individuals (n = 10). RESULTS: The results showed significantly higher mean values for sperm DNA damage, lipid peroxidation and reduced telomere length in spermatozoa of infertile men with previous failed/low fertilization compared with fertile individuals (P < 0.05). CONCLUSIONS: Failed/low fertilization rates could be related to oxidative stress resulting in short telomere length, and also increased sperm chromatin damage and lipid peroxidation. From literature sources, shortened telomere length may lead to detachment of chromosomes from the nuclear membrane, the consequences of which are defects in the process of spermatogenesis, pronuclei formation, and delayed or arrested cell cycle post-ICSI.


Asunto(s)
Infertilidad Masculina/diagnóstico , Infertilidad Masculina/terapia , Inyecciones de Esperma Intracitoplasmáticas , Espermatozoides/patología , Acortamiento del Telómero , Adulto , Compuestos de Boro , Núcleo Celular/metabolismo , Cromatina/metabolismo , Daño del ADN , Fragmentación del ADN , Fertilización , Humanos , Leucocitos/citología , Peroxidación de Lípido , Masculino , Oocitos/metabolismo , Estrés Oxidativo , Protaminas/metabolismo , Telómero/patología
2.
Immunogenetics ; 68(1): 43-54, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26563334

RESUMEN

Multiple sclerosis (MS) is one of the most prevalent autoimmune diseases, which involves the central nervous system. In this illness, Treg/Th17 cell imbalance causes the defect. Several studies revealed that T helper 17 (Th17) cells play a crucial role in pathogenesis, inflammation, and autoimmunity of several autoimmune diseases such as MS. In the present study, we assessed transcript levels of miR-27a and miR-214, in purified CD4+ T cells of MS patients, during relapsing and remitting phases in inducing differentiation of T naïve cells to Th17 cells. Forty RR-MS patient samples including those in relapsing (n=20) and remitting (n=20) phases were participated in this study. In addition, transcript levels of IL-17A, RORγt, IL-23R, Foxp3, and TGF-ß in purified CD4+ T cells of patients in relapsing and remitting phases of RRMS patients were compared to healthy controls. Expression levels of miR-27a and miR-214 were measured by RT-qPCR and compared to healthy control group (n=10). Data indicated upregulation of miR27a in relapsing phase of multiple sclerosis compared to remitting phase and healthy volunteers while miR-214 downregulated in relapsing phase of MS compared to remitting phase and healthy volunteers. In silico studies demonstrated pathways which miR-27a and miR-214 could effect on CD4+ T cell lineage fate including TGF-ß and mTOR signaling, respectively. Our data suggest that miR-27a may probably inhibit negative regulators of Th17 cell differentiation, thus promoting its differentiation while miR-214 has an adverse effect.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , MicroARNs/fisiología , Esclerosis Múltiple Recurrente-Remitente/genética , Células Th17/citología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/genética , Regulación de la Expresión Génica , Humanos , Esclerosis Múltiple Recurrente-Remitente/sangre , Esclerosis Múltiple Recurrente-Remitente/inmunología , Valores de Referencia , Transducción de Señal/genética , Linfocitos T Reguladores/fisiología , Células Th17/inmunología
3.
Cell Biol Int ; 39(5): 629-37, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25572300

RESUMEN

Fndc5 has been recently recognized as a myokine which could be cleaved and secreted into blood stream. It is termed as irisin with an important role in thermogenesis and energy homeostasis. Increased expression of Fndc5 has been reported upon retinoic acid treatment during neural differentiation and its knockdown decreased neural differentiation and neurite outgrowth. This study tries to evaluate the effect of Fndc5 overexpression on rate of neural differentiation in mouse. (Thus, transduced cell line of mouse embryonic stem cell with ability to express Fndc5 under Doxycycline treatment was established. Subsequently, the effect of overexpression of Fndc5 on different stages of neural differentiation was studied). Our study showed an increase enhancement in neuronal precursor markers and mature neuron markers upon overexpression of Fndc5, concluding that Fndc5 facilitates neural differentiation. This effect might be related to increased expression of BDNF following overexpression of Fndc5. Our findings are consistent with recent studies reporting a similar role for Fndc5 in proliferation of neural cells and increase in the expression of neurotrophins like BDNF.


Asunto(s)
Diferenciación Celular/genética , Fibronectinas/genética , Células Madre Embrionarias de Ratones/fisiología , Neurogénesis/genética , Neuronas/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proliferación Celular/genética , Células Cultivadas , Fibronectinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/metabolismo , Neuronas/metabolismo , Regulación hacia Arriba/genética
4.
Hum Mol Genet ; 21(16): 3695-702, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22653751

RESUMEN

To date, mutations in two genes, SPATA16 and DPY19L2, have been identified as responsible for a severe teratozoospermia, namely globozoospermia. The two initial descriptions of the DPY19L2 deletion lead to a very different rate of occurrence of this mutation among globospermic patients. In order to better estimate the contribution of DPY19L2 in globozoospermia, we screened a larger cohort including 64 globozoospermic patients. Twenty of the new patients were homozygous for the DPY19L2 deletion, and 7 were compound heterozygous for both this deletion and a point mutation. We also identified four additional mutated patients. The final mutation load in our cohort is 66.7% (36 out of 54). Out of 36 mutated patients, 69.4% are homozygous deleted, 19.4% heterozygous composite and 11.1% showed a homozygous point mutation. The mechanism underlying the deletion is a non-allelic homologous recombination (NAHR) between the flanking low-copy repeats. Here, we characterized a total of nine breakpoints for the DPY19L2 NAHR-driven deletion that clustered in two recombination hotspots, both containing direct repeat elements (AluSq2 in hotspot 1, THE1B in hotspot 2). Globozoospermia can be considered as a new genomic disorder. This study confirms that DPY19L2 is the major gene responsible for globozoospermia and enlarges the spectrum of possible mutations in the gene. This is a major finding and should contribute to the development of an efficient molecular diagnosis strategy for globozoospermia.


Asunto(s)
Eliminación de Gen , Recombinación Homóloga , Infertilidad Masculina/genética , Proteínas de la Membrana/genética , Homocigoto , Humanos , Desequilibrio de Ligamiento , Masculino , Mutación Puntual , Secuencias Repetitivas de Ácidos Nucleicos
5.
Reprod Biol Endocrinol ; 8: 17, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20178606

RESUMEN

BACKGROUND: Leptin, a 167 amino acid peptide hormone, profoundly effects reproduction exerting its biological effects via interaction with the leptin receptor (ObR) which is widely expressed on peripheral tissues. In this study, we have attempted to assess leptin receptor expression in the spermatozoa of fertile males and those diagnosed with male factor infertility; both at the mRNA or protein levels. METHODS: Semen samples were collected from fertile males and individuals with male factor infertility. In order to evaluate leptin receptor expression several techniques were utilized, including: reverse transcriptase-polymerase chain reaction (RT-PCR), immunostaining, flow cytometry, and western blotting. Mononuclear cells isolated from volunteers' peripheral blood were used as positive controls for leptin receptor expression. RESULTS: leptin receptor was noted on mononuclear cells but we were unable to detect this receptor on spermatozoa at the protein level. Leptin receptor expression was detected on peripheral blood mononuclear cells (PBMCs) as positive controls; however it was not detectable on the spermatozoa of both groups by immunofluorescence microscopy or flow cytometry. Furthermore, positive expression of the ObR long isoform as assessed by RT-PCR was observed in the sperm of only four cases, whereas expression of beta-Actin, a house keeping gene, and HspA2, a testis specific gene, was present in all cases. CONCLUSION: The long isoform of leptin receptor may not be present on human sperm. Species difference may be accounted for diverse reproductive physiology which depends on metabolic requirement. Leptin receptor expression at the mRNA level in some individuals may be related to contamination by other cells in semen.


Asunto(s)
Receptores de Leptina/metabolismo , Espermatozoides/metabolismo , Estudios de Casos y Controles , Expresión Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Leptina/genética , Espermatozoides/patología
6.
Differentiation ; 78(2-3): 59-68, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19616885

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by progressive and irreversible decline of memory. Neuropathological features include the progressive degeneration of cholinergic neurons in the forebrain cholinergic projection system especially nucleus basalis of Meynert (nbM). New cell therapeutic approaches for the replacement of degenerated cells are being researched. The aim of this study was to investigate the production of cholinergic neurons from mouse embryonic stem cells (ESCs) and potential for utilizing ESC-derived neuronal precursor cells (NPCs) and primed NPCs (PNPCs) for cell restorative therapy in a rodent model of AD. NPCs were produced by growth factor-mediated selection under serum-free conditions and differentiated better into cholinergic neurons when NPCs primed with Shh (approximately 22%) in comparison with different cholinergic promoting factors. Behavioral assessment of unilateral nbM ibotenic acid-lesioned rats by Morris water maze and spatial probe test revealed a significant behavioral improvement in memory deficits following transplantation with NPCs and/or PNPCs. Immunohistochemical analysis revealed that the majority (approximately 70%) of the NPCs and/or PNPCs retained neuronal phenotype and approximately 40% of them had a cholinergic cell phenotype following transplantation with no tumor formation, indicating that these may be safe for transplantation. This experimental study has important implications as it suggests that the transplantation of mouse ESC-derived NPCs and/or following commitment to a cholinergic cell phenotype can promote behavioral recovery in a rodent model of AD.


Asunto(s)
Enfermedad de Alzheimer/cirugía , Cognición/fisiología , Células Madre Embrionarias/trasplante , Neuronas/trasplante , Trasplante de Células Madre/métodos , Enfermedad de Alzheimer/fisiopatología , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Agonistas de Aminoácidos Excitadores/farmacología , Técnica del Anticuerpo Fluorescente , Ácido Iboténico/farmacología , Factor Inhibidor de Leucemia/farmacología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Urol J ; 16(5): 488-494, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-30882161

RESUMEN

PURPOSE: The aim of this study was to evaluate expression of Post-Acrosomal WW Binding Protein (PAWP) in infertile men with low and high fertilization post ICSI and also globozoospermic men. MATERIALS AND METHODS: Semen samples were collected from 18 infertile men with previously failed or low fertili-zation (< 25%) post ICSI, 10 men with high fertilization (>50%) post ICSI, 15 globozoospermic men, and 21 fertile individuals. Then, expression of PAWP was assessed at RNA with quantitative Real Time PCR. RESULTS: Relative expression of PAWP in sperm was significantly (P < .05) lower in infertile men with globozo-ospermia (41.5 ± 5.7) or low fertilization rate (43.3±10.4) compared to fertile (138.8 ± 17.3)or men with high fer-tilization (211.6 ± 75.6). In addition, a significant positive correlation (r = 0.628; P = .001) was observed between percentage of fertilization with the relative expression of PAWP. CONCLUSION: Considering solid recent evidences regarding PLC? as the main sperm factor involved in oocyte acti-vation, therefore co-localization of PLC? with PAWP in perinuclear theca may account for the above observation and it is likely that PAWP may have other functions and/or it may assist PLC?.


Asunto(s)
Proteínas Portadoras/análisis , Infertilidad Masculina , Proteínas de Plasma Seminal/análisis , Espermatozoides/química , Adulto , Proteínas Portadoras/biosíntesis , Humanos , Infertilidad Masculina/metabolismo , Masculino , Proteínas de Plasma Seminal/biosíntesis , Espermatozoides/metabolismo
8.
DNA Cell Biol ; 37(4): 373-380, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29461897

RESUMEN

Fibronectin type III domain-containing 5 protein (Fndc5) is responsible for producing a secretory protein termed, "irisin." A modified expression of Fndc5 has been reported in different tissues during development, differentiation processes, as well as in metabolic events such as exercise. One of the important issues to be fixed is whether Fndc5 RNA level and protein content are concerted and modified hand in hand. Therefore, the aim of this study is to assess Fndc5 RNA and protein levels in various tissues of mouse and rat with emphasis on brain. Biopsies from various parts of neonatal and adult mouse and rat tissues were simultaneously assessed for transcript levels of Fndc5 and compared with the respective protein levels at the same time. Data indicated, unlike in muscle, no concerted fluctuations were observed for Fndc5 RNA and protein, especially in brain. Further look at four regions of brain (cerebellum, putamen, hippocampus, and cortex) revealed a similar discrepant expression. To hypothesize whether such discrepancy is arisen by miRNAs, we selected three main miRNAs, which were predicted to target Fndc5 and their expression levels were assessed in central nervous system (CNS) of mouse and hippocampus of rat. miRNA levels showed an antiparallel correlation with protein level of Fndc5, interpreting a putative role in regulating Fndc5 protein content in CNS. This phenomenon may represent the importance of governing Fndc5 content in neural cells, which seems to be crucial for neural function and differentiation.


Asunto(s)
Fibronectinas/análisis , Fibronectinas/genética , Animales , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Fibronectinas/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , Ratas , Ratas Wistar
9.
Iran J Basic Med Sci ; 21(3): 277-283, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29511494

RESUMEN

OBJECTIVES: Multiple sclerosis (MS) is considered as a chronic type of an inflammatory disease characterized by loss of myelin of CNS. Recent evidence indicates that Interleukin 17 (IL-17)-producing T helper cells (Th17 cells) population are increased and regulatory T cells (Treg cells) are decreased in MS. Despite extensive research in understanding the mechanism of Th17 and Treg differentiation, the role of microRNAs in MS is not completely understood. Thereby, as a step closer, we analyzed the expression profile of miR-9-5p and miR-106a-5p, and protein level of retinoic acid receptor (RAR)-related orphan receptor C (RORC; Th17 master transcription factor) as direct target of miR-106a-5p and forkhead box P3 (FOXP3; Treg master transcription factor) as indirect target of miR-9-5p in CD4+ T cells in two groups of relapsing and remitting in our relapsing-remitting MS (RR-MS) patients. MATERIALS AND METHODS: Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was utilized to assess the expression of miRNAs and mRNAs, in 40 RR-MS patients and 11 healthy individuals. Thus, FOXP3 and RAR-related orphan receptor γt (RORγt) was assessed in CD4+T-cells by flow cytometry. We also investigated the role of these miRNAs in Th17/Treg differentiation pathway through bioinformatics tools. RESULTS: An up-regulation of miR-9-5p and down-regulation of miR-106a-5p in relapsing phase of MS patients were observed compared to healthy controls. RORC and FOXP3 were up-regulated in relapsing and remitting phases of MS, respectively. CONCLUSION: Expression pattern of miR-9-5p and miR-106a-5p and their targets suggest a possible inducing role of miR-9-5p and suppressing role of miR-106a-5p in differentiation pathway of Th17 cells during MS pathogenesis.

10.
Int J Fertil Steril ; 10(2): 196-207, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27441053

RESUMEN

BACKGROUND: Globozoospermia is a rare syndrome with an incidence of less than 0.1% among infertile men. Researchers have recently identified a large deletion, about 200 kbp, encompassing the whole length of DPY19L2 or mutations in SPATA16 and PICK1 genes associated with globozoospermia. The aim of this study was to analyze the DPY19L2 gene deletion using polymerase chain reaction technique for the exons 1, 48, 11 and 22 as well as break point (BP) "a" in globozoospermic men. MATERIALS AND METHODS: In this experimental study, genome samples were collected from 27 men with globozoospermia (cases) and 36 fertile individuals (controls), and genomic analysis was carried out on each sample. RESULTS: Deletion of DPY19L2 gene accounted for 74% of individuals with globozoospermia. DPY19L2 gene deletion was considered as the molecular pathogenic factor for the onset of globozoospermia in infertile men. By quantitative real-time polymerase chain reaction (qPCR), we genotyped DPY19L2 deletion and identified carriers within the population. CONCLUSION: This technique may be considered as a method for family counseling and has the potential to be used as a pre-implantation genetic diagnosis, especially in ethnic community with high rate of consanguineous marriages.

11.
Iran J Biotechnol ; 14(3): 169-176, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28959333

RESUMEN

BACKGROUND: Promyelocytic leukemia protein (PML) is a tumor suppressor protein that is involved in myeloid cell differentiation in response to retinoic acid (RA). In addition, RA acts as a natural morphogen in neural development. OBJECTIVES: This study aimed to examine PML gene expression in different stages of in vitro neural differentiation of NT2 cells, and to investigate the possible role of PML in pluripotency and/or neural development. MATERIALS AND METHODS: RA was used as a neural inducer for in vitro neural differentiation of NT2 cells. During this process PML mRNA and protein levels were assessed by quantitative real time RT-PCR (QRT-PCR) and Immunoblotting, respectively. Furthermore bisulfite sequencing PCR (BSP) was used to assess PML promoter methylation in NT2 cells and NT2 derived neuronal precursor cells (NT2.NPCs). RESULTS: QRT-PCR results showed that, PML had maximum expression with significant differences in NT2 derived neuronal precursor cells relative to NT2 cells and NT2 derived neural cells (NT2.NCs). Numerous isoforms of PML with different intensities appeared in immunoblots of pluripotent NT2 cells, NT2.NPCs, and NT2.NCs. Furthermore, the methylation of the PML promoter in NT2.NCs was 2.6 percent lower than NT2 cell. CONCLUSIONS: The observed differences in PML expression in different cellular stages possibly could be attributed to the fact that PML in each developmental state might be involved in different cell signaling machinery and different functions. The appearance of different PML isoforms with more intensity in neural progenitor cells; may suggest apossible role for this protein in neural development.

12.
Cell J ; 18(3): 371-80, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27602319

RESUMEN

OBJECTIVE: MicroRNAs (miRNA) are a class of non-coding RNAs which play key roles in post-transcriptional gene regulation. Previous studies indicate that miRNAs are dysregulated in patients with multiple sclerosis (MS). Th17 and regulatory T (Treg) cells are two subsets of CD4+T-cells which have critical functions in the onset and progression of MS. The current study seeks to distinguish fluctuations in expression of CD4+T-cell derived miR-223 during the relapsing-remitting (RR) phase of MS (RR-MS), as well as the expressions of Th17 and Treg cell markers. MATERIALS AND METHODS: This experimental study used real-time quantitative polymerase chain reaction (qRT-PCR) to evaluate CD4+ T cell derived miR-223 expression patterns in patients that experienced either of the RR-MS phases (n=40) compared to healthy controls (n=12), along with RNA markers for Th17 and Treg cells. We conducted flow cytometry analyses of forkhead box P3 (FOXP3) and RAR-related orphan receptor γt (RORγt) in CD4+T-cells. Putative and validated targets of miR-223 were investigated in the miRWalk and miRTarBase databases, respectively. RESULTS: miR-223 significantly upregulated in CD4+T-cells during the relapsing phase of RR-MS compared to the remitting phase (P=0.000) and healthy individuals (P=0.036). Expression of RORγt, a master transcription factor of Th17, upregulated in the relapsing phase, whereas FOXP3 upregulated in the remitting phase. Additionally, potential targets of miR-223, STAT1, FORKHEAD BOX O (FOXO1) and FOXO3 were predicted by in silico studies. CONCLUSION: miR-223 may have a potential role in MS progression. Therefore, suppression of miR-223 can be proposed as an appropriate approach to control progression of the relapsing phase of MS.

13.
Gene ; 557(2): 123-9, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25497839

RESUMEN

Availability of human embryonic stem cells (hESCs) has enhanced the capability of basic and clinical research in the context of human neural differentiation. Derivation of neural progenitor (NP) cells from hESCs facilitates the process of human embryonic development through the generation of neuronal subtypes. We have recently indicated that fibronectin type III domain containing 5 protein (FNDC5) expression is required for appropriate neural differentiation of mouse embryonic stem cells (mESCs). Bioinformatics analyses have shown the presence of three isoforms for human FNDC5 mRNA. To differentiate which isoform of FNDC5 is involved in the process of human neural differentiation, we have used hESCs as an in vitro model for neural differentiation by retinoic acid (RA) induction. The hESC line, Royan H5, was differentiated into a neural lineage in defined adherent culture treated by RA and basic fibroblast growth factor (bFGF). We collected all cell types that included hESCs, rosette structures, and neural cells in an attempt to assess the expression of FNDC5 isoforms. There was a contiguous increase in all three FNDC5 isoforms during the neural differentiation process. Furthermore, the highest level of expression of the isoforms was significantly observed in neural cells compared to hESCs and the rosette structures known as neural precursor cells (NPCs). High expression levels of FNDC5 in human fetal brain and spinal cord tissues have suggested the involvement of this gene in neural tube development. Additional research is necessary to determine the major function of FDNC5 in this process.


Asunto(s)
Células Madre Embrionarias/metabolismo , Fibronectinas/metabolismo , Línea Celular , Embrión de Mamíferos/metabolismo , Fibronectinas/genética , Expresión Génica , Humanos , Masculino , Neurogénesis , Especificidad de Órganos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
14.
Gene ; 551(2): 127-37, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-25168892

RESUMEN

Fibronectin type III domain-containing 5 protein (Fndc5) is an exercise hormone and its transcript profile in mouse showed high degree of expression in heart, skeletal muscle and brain. Our previous studies indicated a significant increase (approximately 10 fold) in mRNA level of Fndc5 when embryonic stem cells were differentiated into beating bodies. As a step closer to identify the involvement of Fndc5 in the process of cardiomyocyte differentiation, we generated a stably inducible transduced mouse embryonic stem cell (mESC) line that overexpressed Fndc5 following Doxycycline induction. Our results indicated that the overexpression of Fndc5 during spontaneous cardiac differentiation significantly increased not only at RNA levels for mesodermal markers but also at the transcriptional levels for cardiac progenitor and cardiac genes. These data suggest that Fndc5 may be involved in cardiomyocyte differentiation. Therefore, a new hope will be arisen for potential application of this myokine for regeneration of damaged cardiac tissues especially in cardiac failure.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Fibronectinas/genética , Miocitos Cardíacos/metabolismo , Animales , Western Blotting , Línea Celular , Doxiciclina/farmacología , Cuerpos Embrioides/metabolismo , Proteínas Fetales/genética , Fibronectinas/metabolismo , Factores de Transcripción Forkhead/genética , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Miocitos Cardíacos/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/genética , Factores de Tiempo
15.
Avicenna J Med Biotechnol ; 4(4): 160-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23407790

RESUMEN

BACKGROUND: Peroxisome Proliferator Activated Receptor gamma (PPARγ), a member of nuclear receptor superfamily, comprises two isoforms in mouse. These two isoforms are encoded by different mRNAs, which are arisen by alternative promoter usage. There are two promoter regions upstream of PPARγ gene. A 3 kb fragment, containing several transcription factor binding sites, acts as PPARγ1 promoter region. Thus, expression pattern of PPARγ1 isoform is due to the potential transcription factors that could influence its promoter activity. PPARγ, Retinoid X Receptor (RXR) and Vitamin D Receptor (VDR), as nuclear receptors could influence PPARγ gene expression pattern during several differentiation processes. During neural differentiation, PPARγ1 isoform expression reaches to maximal level at neural precursor cell formation. METHODS: A vast computational analysis was carried out to reveal the PPARγ1 promoter region. The putative promoter region was then subcloned upstream of an EGFP reporter gene. Then the functionality of PPARγ1 promoter was assessed in different cell lines. RESULTS: Results indicated that Rosiglitazone increased PPARγ1 promoter regulated EGFP expression of neural precursor cells during Embryoid Body (EB) formation. Furthermore vitamin D reduced PPARγ1 promoter regulated EGFP expression of neural precursor cells during EB formation through binding to its receptor. CONCLUSION: This study suggests that there are potential response elements for PPAR/RXR and VDR/RXR heterodimers in PPARγ1 isoform promoter. Also VDR/RXR heterodimers may decrease PPARγ expression through binding to its promoter.

16.
Int J Dev Biol ; 55(2): 189-95, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21671222

RESUMEN

Stem cells from human exfoliated deciduous teeth (SHED) are highly proliferative, clonogenic and multipotent stem cells with a neural crest cell origin. Additionally, they can be collected with minimal invasiveness in comparison with other sources of mesenchymal stem cells (MSCs). Therefore, SHED could be a desirable option for potential therapeutic applications. In this study, SHEDs were established from enzyme-disaggregated deciduous dental pulp obtained from 6 to 9 year-old children. The cells had typical fibroblastoid morphology and expressed antigens characteristic of MSCs, STRO1, CD146, CD45, CD90, CD106 and CD166, but not the hematopoietic and endothelial markers, CD34 and CD31, as assessed by FACS analysis. Differentiation assessment revealed a strong osteogenic and adipogenic potential of SHEDs. In order to further evaluate the in vitro differentiation potential of SHED into neural cells, a simple short time growth factor-mediated induction was used. Immunofluorescence staining and flow cytometric analysis revealed that SHED rapidly expressed nestin and b-III tubulin, and later expressed intermediate neural markers. In addition, the intensity and percentages of nestin and b-III tubulin and mature neural markers (PSA-NCAM, NeuN, Tau, TH, or GFAP) increased significantly following treatment. Moreover, RT-PCR and Western blot analyses showed that the neural markers were strongly up-regulated after induction. In conclusion, these results provide evidence that SHED can differentiate into neural cells by the expression of a comprehensive set of genes and proteins that define neural-like cells in vitro. SHED cells might be considered as new candidates for the autologous transplantation of a wide variety of neurological diseases and neurotraumatic injuries.


Asunto(s)
Diferenciación Celular , Pulpa Dental/citología , Células Madre Mesenquimatosas/citología , Neuronas/citología , Diente Primario/citología , Biomarcadores , Western Blotting , Proliferación Celular , Células Cultivadas , Niño , Pulpa Dental/embriología , Humanos , Proteínas de Filamentos Intermediarios/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Nestina , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa , Exfoliación Dental , Tubulina (Proteína)/biosíntesis
17.
Stem Cells Dev ; 18(2): 259-67, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18422402

RESUMEN

Understanding neuroectoderm formation and its subsequent diversification to functional neural subtypes remains elusive. We have shown here for the first time that embryonic stem cells (ESCs) can differentiate into neurons and motor neurons (MNs) by using a coculture embryonic notochord model in vitro. Mouse ESCs were induced to form neural precursors via timed exposure to retinoic acid (RA) using the 4-/4+ RA protocol. These cells were then cocultured with alginate bead-encapsulated notochords isolated from Hamburger and Hamilton stage 6-10 chick embryos. The use of notochord alone was not able to induce neural differentiation from ESCs, and, therefore, notochord does not possess neural inducing activity. Hence, the most successful neuronal cells and MN differentiation was only observed following the coculture of RA-pretreated ESCs with notochord. This resulted in a significantly greater number of cells expressing microtubule-associated protein-2 (MAP2), HB9, choline acetyltransferase (ChAT) and MN-specific genes. While further characterization of these differentiated cells will be essential before transplantation studies commence, these data illustrate the effectiveness of embryonic notochord coculture in providing valuable molecular cues for directed differentiation of ESCs toward an MN lineage.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Neuronas Motoras/citología , Neuronas Motoras/efectos de los fármacos , Notocorda/citología , Notocorda/efectos de los fármacos , Tretinoina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Pollos , Técnicas de Cocultivo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones
18.
Toxicol In Vitro ; 23(7): 1422-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19596060

RESUMEN

Embryonic stem cell (ESC)-derived beating cardiomyocytes may be considered as a suitable model for in vitro assessment of pharmacological and toxicological studies. In this model, laboratory animals are not required. In addition, physiological functions, such as heart beat, are assessed rather than single parameters such as cell viability. Here we report that doxorubicin (DOX) cardiotoxicity on mouse ESC-derived beating cardiomyocytes can be ameliorated by treatment with dexamethasone (DEX) when DEX is administrated only before DOX and not in combination with DOX. DEX effect appears to be mediated via glucocorticoid receptor and increases cardiomyocyte-specific gene expression. Cardiotoxicity of DOX can be augmented by calcium channel blocker, verapamil (VER) which also decreases the expression of cardiac gene markers. This model provides us with a clinical suggestion which proposes that the beneficial effect of DEX is obtained when DEX was added before DOX administration.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Dexametasona/farmacología , Doxorrubicina/toxicidad , Células Madre Embrionarias/citología , Glucocorticoides/farmacología , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Citotoxinas/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Ratones , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Verapamilo/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA