Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 386(20): 1889-1898, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35584156

RESUMEN

BACKGROUND: Xenografts from genetically modified pigs have become one of the most promising solutions to the dearth of human organs available for transplantation. The challenge in this model has been hyperacute rejection. To avoid this, pigs have been bred with a knockout of the alpha-1,3-galactosyltransferase gene and with subcapsular autologous thymic tissue. METHODS: We transplanted kidneys from these genetically modified pigs into two brain-dead human recipients whose circulatory and respiratory activity was maintained on ventilators for the duration of the study. We performed serial biopsies and monitored the urine output and kinetic estimated glomerular filtration rate (eGFR) to assess renal function and xenograft rejection. RESULTS: The xenograft in both recipients began to make urine within moments after reperfusion. Over the 54-hour study, the kinetic eGFR increased from 23 ml per minute per 1.73 m2 of body-surface area before transplantation to 62 ml per minute per 1.73 m2 after transplantation in Recipient 1 and from 55 to 109 ml per minute per 1.73 m2 in Recipient 2. In both recipients, the creatinine level, which had been at a steady state, decreased after implantation of the xenograft, from 1.97 to 0.82 mg per deciliter in Recipient 1 and from 1.10 to 0.57 mg per deciliter in Recipient 2. The transplanted kidneys remained pink and well-perfused, continuing to make urine throughout the study. Biopsies that were performed at 6, 24, 48, and 54 hours revealed no signs of hyperacute or antibody-mediated rejection. Hourly urine output with the xenograft was more than double the output with the native kidneys. CONCLUSIONS: Genetically modified kidney xenografts from pigs remained viable and functioning in brain-dead human recipients for 54 hours, without signs of hyperacute rejection. (Funded by Lung Biotechnology.).


Asunto(s)
Rechazo de Injerto , Trasplante de Riñón , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/cirugía , Muerte Encefálica , Rechazo de Injerto/etiología , Rechazo de Injerto/patología , Rechazo de Injerto/prevención & control , Xenoinjertos/trasplante , Humanos , Riñón/patología , Riñón/fisiología , Trasplante de Riñón/efectos adversos , Trasplante de Riñón/métodos , Porcinos/cirugía , Trasplante Heterólogo/efectos adversos , Trasplante Heterólogo/métodos
2.
Am J Transplant ; 22(4): 1037-1053, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35049121

RESUMEN

A radical solution is needed for the organ supply crisis, and the domestic pig is a promising organ source. In preparation for a clinical trial of xenotransplantation, we developed an in vivo pre-clinical human model to test safety and feasibility tenets established in animal models. After performance of a novel, prospective compatible crossmatch, we performed bilateral native nephrectomies in a human brain-dead decedent and subsequently transplanted two kidneys from a pig genetically engineered for human xenotransplantation. The decedent was hemodynamically stable through reperfusion, and vascular integrity was maintained despite the exposure of the xenografts to human blood pressure. No hyperacute rejection was observed, and the kidneys remained viable until termination 74 h later. No chimerism or transmission of porcine retroviruses was detected. Longitudinal biopsies revealed thrombotic microangiopathy that did not progress in severity, without evidence of cellular rejection or deposition of antibody or complement proteins. Although the xenografts produced variable amounts of urine, creatinine clearance did not recover. Whether renal recovery was impacted by the milieu of brain death and/or microvascular injury remains unknown. In summary, our study suggests that major barriers to human xenotransplantation have been surmounted and identifies where new knowledge is needed to optimize xenotransplantation outcomes in humans.


Asunto(s)
Rechazo de Injerto , Riñón , Animales , Animales Modificados Genéticamente , Rechazo de Injerto/patología , Xenoinjertos , Humanos , Estudios Prospectivos , Porcinos , Trasplante Heterólogo
3.
Bioinformatics ; 31(8): 1191-8, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25481010

RESUMEN

RATIONALE: The growing recognition of the importance of splicing, together with rapidly accumulating RNA-sequencing data, demand robust high-throughput approaches, which efficiently analyze experimentally derived whole-transcriptome splice profiles. RESULTS: We have developed a computational approach, called SNPlice, for identifying cis-acting, splice-modulating variants from RNA-seq datasets. SNPlice mines RNA-seq datasets to find reads that span single-nucleotide variant (SNV) loci and nearby splice junctions, assessing the co-occurrence of variants and molecules that remain unspliced at nearby exon-intron boundaries. Hence, SNPlice highlights variants preferentially occurring on intron-containing molecules, possibly resulting from altered splicing. To illustrate co-occurrence of variant nucleotide and exon-intron boundary, allele-specific sequencing was used. SNPlice results are generally consistent with splice-prediction tools, but also indicate splice-modulating elements missed by other algorithms. SNPlice can be applied to identify variants that correlate with unexpected splicing events, and to measure the splice-modulating potential of canonical splice-site SNVs. AVAILABILITY AND IMPLEMENTATION: SNPlice is freely available for download from https://code.google.com/p/snplice/ as a self-contained binary package for 64-bit Linux computers and as python source-code. CONTACT: pmudvari@gwu.edu or horvatha@gwu.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Intrones/genética , Empalme del ARN/genética , Epitelio Pigmentado de la Retina/metabolismo , Análisis de Secuencia de ARN/métodos , Programas Informáticos , Células Cultivadas , Exones/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Neoplasias/genética , ARN/genética , Epitelio Pigmentado de la Retina/citología
4.
J Neurosci ; 33(41): 16346-59, 2013 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-24107965

RESUMEN

The retinal pigment epithelium (RPE) is a highly specialized CNS tissue that plays crucial roles in retinal homeostasis. Age-related morphological changes in the RPE have been associated with retinal degenerative disorders; our understanding of the underlying molecular mechanisms, however, remains incomplete. Here we report on a key role of Klotho (Kl), an aging-suppressor gene, in retinal health and RPE physiology. Kl(-/-) mice show RPE and photoreceptor degeneration, reduced pigment synthesis in the RPE, and impaired phagocytosis of the outer segment of the photoreceptors. Klotho protein (KL) is expressed in primary cultured human RPE, and regulates pigment synthesis by increasing the expression of MITF (microphthalmia transcription factor) and TYR (tyrosinase), two pivotal genes in melanogenesis. Importantly, KL increases phagocytosis in cultured RPE by inducing gene expression of MERTK/AXL/TYRO3. These effects of KL are mediated through cAMP-PKA-dependent phosphorylation of transcription factor CREB. In cultured human RPE, KL increases the l-3,4-dihydroxyphenylalanine synthesis and inhibits vascular endothelial growth factor (VEGF) secretion from basal membrane by inhibiting IGF-1 signaling and VEGF receptor 2 phosphorylation. KL also regulates the expression of stress-related genes in RPE, lowers the production of reactive oxygen species, and thereby, protects RPE from oxidative stress. Together, our results demonstrate a critical function for KL in mouse retinal health in vivo, and a protective role toward human RPE cells in vitro. We conclude that KL is an important regulator of RPE homeostasis, and propose that an age-dependent decline of KL expression may contribute to RPE degeneration and retinal pathology.


Asunto(s)
Glucuronidasa/metabolismo , Estrés Oxidativo/fisiología , Epitelio Pigmentado de la Retina/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica/fisiología , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Proteínas Klotho , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Degeneración Retiniana/metabolismo , Transducción de Señal/fisiología
5.
Stem Cells ; 31(10): 2205-17, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23836497

RESUMEN

Studies on spermatogonial stem cells (SSCs) are of unusual significance because they are the unique stem cells that transmit genetic information to subsequent generations and they can acquire pluripotency to become embryonic stem-like cells that have therapeutic applications in human diseases. MicroRNAs (miRNAs) have recently emerged as critical endogenous regulators in mammalian cells. However, the function and mechanisms of individual miRNAs in regulating SSC fate remain unknown. Here, we report for the first time that miRNA-20 and miRNA-106a are preferentially expressed in mouse SSCs. Functional assays in vitro and in vivo using miRNA mimics and inhibitors reveal that miRNA-20 and miRNA-106a are essential for renewal of SSCs. We further demonstrate that these two miRNAs promote renewal at the post-transcriptional level via targeting STAT3 and Ccnd1 and that knockdown of STAT3, Fos, and Ccnd1 results in renewal of SSCs. This study thus provides novel insights into molecular mechanisms regulating renewal and differentiation of SSCs and may have important implications for regulating male reproduction.


Asunto(s)
Células Madre Adultas/fisiología , Ciclina D1/genética , MicroARNs/fisiología , Interferencia de ARN , Factor de Transcripción STAT3/genética , Animales , Secuencia de Bases , Proliferación Celular , Células Cultivadas , Ciclina D1/metabolismo , Femenino , Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Familia de Multigenes , Fenotipo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Factor de Transcripción STAT3/metabolismo , Espermatogénesis/genética , Testículo/citología
6.
Nat Med ; 29(8): 1989-1997, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37488288

RESUMEN

Genetically modified xenografts are one of the most promising solutions to the discrepancy between the numbers of available human organs for transplantation and potential recipients. To date, a porcine heart has been implanted into only one human recipient. Here, using 10-gene-edited pigs, we transplanted porcine hearts into two brain-dead human recipients and monitored xenograft function, hemodynamics and systemic responses over the course of 66 hours. Although both xenografts demonstrated excellent cardiac function immediately after transplantation and continued to function for the duration of the study, cardiac function declined postoperatively in one case, attributed to a size mismatch between the donor pig and the recipient. For both hearts, we confirmed transgene expression and found no evidence of cellular or antibody-mediated rejection, as assessed using histology, flow cytometry and a cytotoxic crossmatch assay. Moreover, we found no evidence of zoonotic transmission from the donor pigs to the human recipients. While substantial additional work will be needed to advance this technology to human trials, these results indicate that pig-to-human heart xenotransplantation can be performed successfully without hyperacute rejection or zoonosis.


Asunto(s)
Anticuerpos , Rechazo de Injerto , Animales , Humanos , Porcinos , Trasplante Heterólogo/métodos , Xenoinjertos , Corazón , Animales Modificados Genéticamente
7.
Stem Cells ; 29(5): 825-35, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21480547

RESUMEN

Age-related macular degeneration (AMD) is one of the major causes of blindness in aging population that progresses with death of retinal pigment epithelium (RPE) and photoreceptor degeneration inducing impairment of central vision. Discovery of human induced pluripotent stem (hiPS) cells has opened new avenues for the treatment of degenerative diseases using patient-specific stem cells to generate tissues and cells for autologous cell-based therapy. Recently, RPE cells were generated from hiPS cells. However, there is no evidence that those hiPS-derived RPE possess specific RPE functions that fully distinguish them from other types of cells. Here, we show for the first time that RPE generated from hiPS cells under defined conditions exhibit ion transport, membrane potential, polarized vascular endothelial growth factor secretion, and gene expression profile similar to those of native RPE. The hiPS-RPE could therefore be a very good candidate for RPE replacement therapy in AMD. However, these cells show rapid telomere shortening, DNA chromosomal damage, and increased p21 expression that cause cell growth arrest. This rapid senescence might affect the survival of the transplanted cells in vivo and therefore, only the very early passages should be used for regeneration therapies. Future research needs to focus on the generation of "safe" as well as viable hiPS-derived somatic cells.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Transporte Iónico/fisiología , Potenciales de la Membrana/fisiología , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Daño del ADN/genética , Electrofisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Immunoblotting , Transporte Iónico/genética , Potenciales de la Membrana/genética , Fagocitosis/genética , Fagocitosis/fisiología , Reacción en Cadena de la Polimerasa
8.
Biol Reprod ; 82(2): 363-72, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19846602

RESUMEN

This study was designed to isolate, characterize, and culture human spermatogonia. Using immunohistochemistry on tubule sections, we localized GPR125 to the plasma membrane of a subset of the spermatogonia. Immunohistochemistry also showed that MAGEA4 was expressed in all spermatogonia (A(dark), A(pale), and type B) and possibly preleptotene spermatocytes. Notably, KIT was expressed in late spermatocytes and round spermatids, but apparently not in human spermatogonia. UCHL1 was found in the cytoplasm of spermatogonia, whereas POU5F1 was not detected in any of the human germ cells. GFRA1 and ITGA6 were localized to the plasma membrane of the spermatogonia. Next, we isolated GPR125-positive spermatogonia from adult human testes using a two-step enzymatic digestion followed by magnetic-activated cell sorting. The isolated GPR125-positive cells coexpressed GPR125, ITGA6, THY1, and GFRA1, and they could be cultured for short periods of time and exhibited a marked increase in cell numbers as shown by a proliferation assay. Immunocytochemistry of putative stem cell genes after 2 wk in culture revealed that the cells were maintained in an undifferentiated state. MAPK1/3 phosphorylation was increased after 2 wk of culture of the GPR125-positive spermatogonia compared to the freshly isolated cells. Taken together, these results indicate that human spermatogonia share some but not all phenotypes with spermatogonial stem cells (SSCs) and progenitors from other species. GPR125-positive spermatogonia are phenotypically putative human SSCs and retain an undifferentiated status in vitro. This study provides novel insights into the molecular characteristics, isolation, and culture of human SSCs and/or progenitors and suggests that the MAPK1/3 pathway is involved in their proliferation.


Asunto(s)
Espermatogonias/citología , Testículo/citología , Adolescente , Adulto , Antígenos de Neoplasias/análisis , Biomarcadores/análisis , Técnicas de Cultivo de Célula , División Celular , Membrana Celular/química , Separación Celular , Activación Enzimática , Humanos , Inmunohistoquímica , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas de Neoplasias/análisis , Fenotipo , Receptores Acoplados a Proteínas G/análisis , Espermátides/química , Espermatocitos/química , Espermatogonias/química , Espermatogonias/clasificación , Células Madre/química , Células Madre/citología
9.
Reproduction ; 139(6): 1011-20, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20371641

RESUMEN

Spermatogenesis in man starts with spermatogonial stem cells (SSCs), and leads to the production of sperm in approximately 64 days, common to old and young men. Sperm from elderly men are functional and able to fertilize eggs and produce offspring, even though daily sperm production is more than 50% lower and damage to sperm DNA is significantly higher in older men than in those who are younger. Our hypothesis is that the SSC/spermatogonial progenitors themselves age. To test this hypothesis, we studied the gene expression profile of mouse SSC/progenitor cells at several ages using microarrays. After sequential enzyme dispersion, we purified the SSC/progenitors with immunomagnetic cell sorting using an antibody to GFRA1, a known SSC/progenitor cell marker. RNA was isolated and used for the in vitro synthesis of amplified and labeled cRNAs that were hybridized to the Affymetrix mouse genome microarrays. The experiments were repeated twice with different cell preparations, and statistically significant results are presented. Quantitative RT-PCR analysis was used to confirm the microarray results. Comparison of four age groups (6 days, 21 days, 60 days, and 8 months old) showed a number of genes that were expressed specifically in the older mice. Two of them (i.e. Icam1 and Selp) have also been shown to mark aging hematopoietic stem cells. On the other hand, the expression levels of the genes encoding the SSC markers Gfra1 and Plzf did not seem to be significantly altered by age, indicating that age affects only certain SSC/progenitor properties.


Asunto(s)
Envejecimiento/genética , Expresión Génica/genética , Espermatogonias/metabolismo , Células Madre/metabolismo , Animales , Recuento de Células , Senescencia Celular/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/análisis , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/metabolismo , Separación Inmunomagnética , Molécula 1 de Adhesión Intercelular/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Selenoproteína P/genética , Espermatogonias/química , Espermatogonias/citología , Células Madre/química , Células Madre/citología , Testículo/citología
10.
Stem Cells ; 27(10): 2580-90, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19688838

RESUMEN

Spermatogenesis is the process that involves the division and differentiation of spermatogonial stem cells into spermatozoa. However, the autocrine molecules and signaling pathways controlling their fate remain unknown. This study was designed to identify novel growth factors and signaling pathways that regulate proliferation, differentiation, and survival of spermatogonial stem/progenitor cells. To this end, we have for the first time explored the expression, function, and signaling pathway of Nodal, a member of the transforming growth factor-beta superfamily, in mouse spermatogonial stem/progenitor cells. We demonstrate that both Nodal and its receptors are present in these cells and in a spermatogonial stem/progenitor cell line (C18-4 cells), whereas Nodal is undetected in Sertoli cells or differentiated germ cells, as assayed by reverse transcription-polymerase chain reaction, Western blots, and immunocytochemistry. Nodal promotes proliferation of spermatogonial stem/progenitor cells and C18-4 cells, whereas Nodal receptor inhibitor SB431542 blocks their propagation as shown by proliferation and bromodeoxyuridine incorporation assays. Nodal knockdown by RNA interference results in a marked increase of cell apoptosis and a reduction of cell division as indicated by terminal deoxynucleotidyl transferase dUTP nick-end labeling and proliferation assays. Conversely, overexpression of Nodal leads to an increase of cell proliferation. Nodal activates Smad2/3 phosphorylation, Oct-4 transcription, cyclin D1, and cyclin E expression, whereas SB431542 completely abolishes their increase. Together, Nodal was identified as the first autocrine signaling molecule that promotes proliferation of mouse spermatogonial stem/progenitor cells via Smad2/3 and Oct-4 activation. This study thus provides novel and important insights into molecular mechanisms regulating proliferation and survival of spermatogonial stem/progenitor cells.


Asunto(s)
Proteína Nodal/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteína Smad2/metabolismo , Espermatogonias/metabolismo , Células Madre/metabolismo , Animales , Apoptosis/genética , Comunicación Autocrina/fisiología , Benzamidas/farmacología , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular , Supervivencia Celular/fisiología , Ciclinas/metabolismo , Dioxoles/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteína Nodal/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Proteína Smad2/genética , Espermatogénesis/fisiología , Espermatogonias/citología , Células Madre/citología
11.
Birth Defects Res C Embryo Today ; 87(1): 27-34, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19306345

RESUMEN

Spermatogonial stem cells (SSCs) have unique characteristics in that they produce sperm that transmit genetic information from generation to generation and they can be reprogrammed spontaneously to form embryonic stem (ES)-like cells to acquire pluripotency. In rodents, it is generally believed that the A-single (A(s)) is the stem cell population, whereas the A-paired (A(pr)) and A-aligned (A(al)) represent the progenitor spermatogonial population. The A(1) to A(4) cells, intermediate, and type B spermatogonia are considered differentiated spermatogonia. In human, very little information is available about SSCs, except for the earlier work of Clermont and colleagues who demonstrated that there are two different types of A spermatogonia, the A(dark) and A(pale) spermatogonia. The A(dark) spermatogonia were referred to as the reserve stem cells, whereas the A(pale) were considered the renewing stem cells. In this review, we outline several spermatogonial renewal schemes for both rodents and primates, including man. We also compare phenotypic markers for spermatogonia/spermatogonial stem cells in rodents and humans and address SSC potential and therapeutic application.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Pluripotentes/fisiología , Espermatogonias/fisiología , Animales , Biomarcadores/metabolismo , Humanos , Masculino
12.
Reproduction ; 137(6): 901-11, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19318589

RESUMEN

Small RNA molecules (small RNAs), including small interfering RNAs (siRNAs), microRNAs (miRNAs), and piwi-interacting RNAs (piRNAs), have recently emerged as important regulators of gene expression at the post-transcriptional or translation level. Significant progress has recently been made utilizing small RNAs in elucidating the molecular mechanisms regulating spermatogenesis. Spermatogenesis is a complex process that involves the division and eventual differentiation of spermatogonial stem cells into mature spermatozoa. The process of spermatogenesis is composed of several phases: mitotic proliferation of spermatogonia to produce spermatocytes; two meiotic divisions of spermatocytes to generate haploid round spermatids; and spermiogenesis, the final phase that involves the maturation of early-round spermatids into elongated mature spermatids. A number of miRNAs are expressed abundantly in male germ cells throughout spermatogenesis, while piRNAs are only present in pachytene spermatocytes and round spermatids. In this review, we first address the synthesis, mechanisms of action, and functions of siRNA, miRNA, and piRNA, and then we focus on the recent advancements in defining the small RNAs in the regulation of spermatogenesis. Concerns pertaining to the use of siRNAs in exploring spermatogenesis mechanisms and open questions in miRNAs and piRNAs in this field are highlighted. The potential applications of small RNAs to male contraception and treatment for male infertility and testicular cancer are also discussed.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , ARN Interferente Pequeño/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Animales , Anticoncepción , Terapia Genética , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/terapia , Masculino , MicroARNs/uso terapéutico , ARN Interferente Pequeño/uso terapéutico , Neoplasias Testiculares/genética , Neoplasias Testiculares/terapia
13.
Reproduction ; 138(1): 151-62, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19419993

RESUMEN

Spermatogonial stem cells (SSCs) self-renew throughout life to produce progenitor cells that are able to differentiate into spermatozoa. However, the mechanisms underlying the cell fate determination between self-renewal and differentiation have not yet been delineated. Culture conditions and growth factors essential for self-renewal and proliferation of mouse SSCs have been investigated, but no information is available related to growth factors that affect fate determination of human spermatogonia. Wnts form a large family of secreted glycoproteins, the members of which are involved in cell proliferation, differentiation, organogenesis, and cell migration. Here, we show that Wnts and their receptors Fzs are expressed in mouse spermatogonia and in the C18-4 SSC line. We demonstrate that WNT3A induces cell proliferation, morphological changes, and cell migration in C18-4 cells. Furthermore, we show that beta-catenin is activated during testis development in 21-day-old mice. In addition, our study demonstrates that WNT3A sustained adult human embryonic stem (ES)-like cells derived from human germ cells in an undifferentiated stage, expressing essential human ES cell transcription factors. These results demonstrate for the first time that Wnt/beta-catenin pathways, especially WNT3A, may play an important role in the regulation of mouse and human spermatogonia.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Transducción de Señal , Espermatogonias/metabolismo , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Movimiento Celular , Forma de la Célula , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Proteínas Dishevelled , Receptores Frizzled/metabolismo , Genes Reporteros , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Fosfoproteínas/metabolismo , Fosforilación , Transfección , Proteína Wnt3A/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
14.
Stem Cells ; 26(1): 266-78, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17962702

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) plays a crucial role in regulating the proliferation of spermatogonial stem cells (SSC). The signaling pathways mediating the function of GDNF in SSC remain unclear. This study was designed to determine whether GDNF signals via the Ras/ERK1/2 pathway in the C18-4 cells, a mouse SSC line. The identity of this cell line was confirmed by the expression of various markers for germ cells, proliferating spermatogonia, and SSC, including GCNA1, Vasa, Dazl, PCNA, Oct-4, GFRalpha1, Ret, and Plzf. Western blot analysis revealed that GDNF activated Ret tyrosine phosphorylation. All 3 isoforms of Shc were phosphorylated upon GDNF stimulation, and GDNF induced the binding of the phosphorylated Ret to Shc and Grb2 as indicated by immunoprecipitation and Western blotting. The active Ras was induced by GDNF, which further activated ERK1/2 phosphorylation. GDNF stimulated the phosphorylation of CREB-1, ATF-1, and CREM-1, and c-fos transcription. Notably, the increase in ERK1/2 phosphorylation, c-fos transcription, bromodeoxyuridine incorporation, and metaphase counts induced by GDNF, was completely blocked by pretreatment with PD98059, a specific inhibitor for MEK1, the upstream regulator of ERK1/2. GDNF stimulation eventually upregulated cyclin A and CDK2 expression. Together, these data suggest that GDNF induces CREB/ATF-1 family member phosphorylation and c-fos transcription via the Ras/ERK1/2 pathway to promote the proliferation of SSC. Unveiling GDNF signaling cascades in SSC has important implications in providing attractive targets for male contraception as well as for the regulation of stem cell renewal vs. differentiation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Espermatozoides/citología , Células Madre/citología , Proteínas ras/metabolismo , Animales , Western Blotting , Diferenciación Celular , Línea Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Expresión Génica , Regulación de la Expresión Génica , Inmunoprecipitación , Masculino , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-fos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Espermatozoides/metabolismo , Células Madre/metabolismo
15.
Biochim Biophys Acta ; 1574(3): 375-82, 2002 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-11997106

RESUMEN

The Drosophila neuralized (neur) gene belongs to the neurogenic group of genes involved in regulating cell-cell interactions required for neural precursor development. neur mutant phenotypes include strong overcommitment to neural fates at the expense of epidermal fates. The human neuralized homolog (NEURL) has been recently determined and found to map to chromosome 10q25.1 within the region frequently deleted in malignant astrocytomas. Because of its potential importance in developmental processes, we analyzed the structure of the mouse homolog, Neurl, and its expression pattern in embryonic tissues. Neurl activity is detected from early developmental stages in several tissues and organs including neural tissues, limbs, the skeletal system, sense organs and internal organs undergoing epithelial-mesenchymal interactions. Neurl encodes a polypeptide associated with the plasma membrane but also detected in the cytoplasm. Similarly to the Drosophila gene, mammalian neuralized may code for an important regulatory factor.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Plexo Coroideo/química , Plexo Coroideo/embriología , Mapeo Cromosómico , Clonación Molecular , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Hígado/química , Hígado/embriología , Pulmón/química , Pulmón/embriología , Mesencéfalo/química , Mesencéfalo/embriología , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/metabolismo , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Transfección
16.
FEBS J ; 281(23): 5251-64, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25251993

RESUMEN

MicroRNA 184 (miR-184) is known to play a key role in neurological development and apoptosis and is highly expressed in mouse brain, mouse corneal epithelium, zebrafish lens and human retinal pigment epithelium (RPE). However, the role of miR-184 in RPE is largely unknown. We investigated the role of miR-184 in RPE and its possible implication in age-related macular degeneration (AMD). Proteomic analysis identified the ezrin (EZR) gene as a target of miR-184 in human RPE. EZR is a membrane cytoskeleton crosslinker that is also known to bind to lysosomal-associated membrane protein 1 (LAMP-1) during the formation of phagocytic vacuoles. In adult retinal pigment epithelium 19 (ARPE19) cells, inhibition of miR-184 resulted in upregulation of EZR mRNA and EZR protein, and induced downregulation of LAMP-1. The inhibition of miR-184 decreased EZR-bound LAMP-1 protein levels and affected phagocytic activity in ARPE19 cells. In primary culture of human RPE isolated from eyes of AMD donors (AMD RPE), miR-184 was significantly downregulated compared with control (normal) RPE. Downregulation of miR-184 was consistent with significantly lower levels of LAMP-1 protein in AMD RPE, and overexpression of MIR-184 in AMD RPE was able to rescue LAMP-1 protein expression to normal levels. Altogether, these observations suggest a novel role for miR-184 in RPE health and support a model proposing that downregulation of miR-184 expression during aging may result in dysregulation of RPE function, contributing to retinal degeneration.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Proteínas de Membrana de los Lisosomas/fisiología , Degeneración Macular/etiología , MicroARNs/fisiología , Fagocitosis , Epitelio Pigmentado de la Retina/metabolismo , Adolescente , Anciano , Anciano de 80 o más Años , Células Cultivadas , Niño , Proteínas del Citoesqueleto/genética , Regulación hacia Abajo , Femenino , Humanos , Proteínas de Membrana de los Lisosomas/genética , Degeneración Macular/genética , Degeneración Macular/metabolismo , Masculino , Persona de Mediana Edad , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo
17.
Cell Rep ; 3(6): 2179-90, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23791531

RESUMEN

Understanding the extent of genomic transcription and its functional relevance is a central goal in genomics research. However, detailed genome-wide investigations of transcriptome complexity in major mammalian organs have been scarce. Here, using extensive RNA-seq data, we show that transcription of the genome is substantially more widespread in the testis than in other organs across representative mammals. Furthermore, we reveal that meiotic spermatocytes and especially postmeiotic round spermatids have remarkably diverse transcriptomes, which explains the high transcriptome complexity of the testis as a whole. The widespread transcriptional activity in spermatocytes and spermatids encompasses protein-coding and long noncoding RNA genes but also poorly conserves intergenic sequences, suggesting that it may not be of immediate functional relevance. Rather, our analyses of genome-wide epigenetic data suggest that this prevalent transcription, which most likely promoted the birth of new genes during evolution, is facilitated by an overall permissive chromatin in these germ cells that results from extensive chromatin remodeling.


Asunto(s)
ARN/genética , Testículo/fisiología , Transcripción Genética , Transcriptoma , Animales , Evolución Biológica , Humanos , Masculino , Mamíferos , Espermatocitos/citología , Espermatocitos/fisiología , Testículo/citología
18.
Methods Mol Biol ; 825: 45-57, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22144235

RESUMEN

Mammalian spermatogenesis is a process whereby male germ-line stem cells (spermatogonial stem cells) divide and differentiate into sperm. Although a great deal of progress has been made in the isolation and characterization of spermatogonial stem cells (SSCs) in rodents, little is known about human SSCs. We have recently isolated human G protein-coupled receptor 125 (GPR125)-positive spermatogonia and GDNF family receptor alpha 1 (GFRA1)-positive spermatogonia using a 2-step enzymatic digestion and magnetic-activated cell sorting (MACS) from adult human testes. Cell purities of isolated human GPR125- and GFRA1-positive spermatogonia after MACS are greater than 95%, and cell viability is over 96%. The isolated GPR125- and GFRA1-positive spermatogonia coexpress GPR125, integrin, alpha 6 (ITGA6), THY1 (also known as CD90), GFRA1, and ubiquitin carboxyl-terminal esterase L1 (UCHL1), markers for rodent or pig SSCs/progenitors, suggesting that GPR125- and GFRA1-positive spermatogonia are phenotypically the SSCs in human testis. Human GPR125-positive spermatogonia can be cultured for 2 weeks with a remarkable increase in cell number. Immunocytochemistry further reveals that GPR125-positive spermatogonia can be maintained in an undifferentiated state in vitro. Collectively, the methods using enzymatic digestion and MACS can efficiently isolate and purify SSCs from adult human testis with consistent and high quality. The ability of isolating and characterizing human SSCs could provide a population of stem cells with high purity for mechanistic studies on human SSC self-renewal and differentiation as well as potential applications of human SSCs in regenerative medicine.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Espermatogonias/citología , Células Madre/citología , Adulto , Diferenciación Celular , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Inmunohistoquímica/métodos , Masculino , Receptores Acoplados a Proteínas G/metabolismo , Espermatogonias/metabolismo , Células Madre/metabolismo , Testículo/citología , Donantes de Tejidos
19.
J Stem Cell Res Ther ; 2(2)2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24466499

RESUMEN

Recently we and two other groups have shown that human spermatogonial stem cells (SSCs) have the potential to become pluripotent in vitro in defined culture conditions and to differentiate into cells of the three embryonic germ layers. This discovery could open new avenues for autologous cell-based therapy in degenerative diseases, bypassing the ethical and immunological problems related to the human embryonic stem cells. In addition, human SSCs could be used to treat infertility in cancer survival children. However, in order to reprogram SSCs into pluripotency, or to preserve them for repopulation of infertile testes, the first and limiting step is to have access to a highly purified human SSC population that could be multiplied and efficiently cultured in vitro maintaining their molecular and cellular characteristics. Although various studies have attempted to identify molecular markers of human SSCs, to date there is still limited information related to the specific markers that could be used for their isolation and optimized purification that allows long-term in vitro culture of isolated human SSCs. Here using SSEA-4 as an optimal marker for isolation of a subpopulation of SSCs, we show that SSEA-4 positive cells express the highest level of SSC genes compared to other subpopulations isolated with different markers, and can be maintained in culture for over 14 passages which we were unable to obtain with other SSCs markers including GPR125 and ITGA6. In addition, we have established a new technology for cell sorting and long-term culture of human SSC-SSEA-4 positive cells that maximizes the purity and viability of the sorted cells. Our findings are crucial and could be used for the most efficient isolation, purification and long-term culture of SSCs for clinical applications in regenerative medicine, or for preparation of human SSCs for autologous treatment of infertility in cancer survival children.

20.
Microsc Res Tech ; 72(8): 586-95, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19263492

RESUMEN

Spermatogenesis is the process that involves the division and differentiation of spermatogonial stem cells (SSCs) into mature spermatozoa. SSCs are a subpopulation of type A spermatogonia resting on the basement membrane in the mammalian testis. Self-renewal and differentiation of SSCs are the foundation of normal spermatogenesis, and thus a better understanding of molecular mechanisms and signaling pathways in the SSCs is of paramount importance for the regulation of spermatogenesis and may eventually lead to novel targets for male contraception as well as for gene therapy of male infertility and testicular cancer. Uncovering the molecular mechanisms is also of great interest to a better understanding of SSC aging and for developing novel therapeutic strategies for degenerative diseases in view of the recent work demonstrating the pluripotent potential of the SSC. Progress has recently been made in elucidating the signaling molecules and pathways that determine cell fate decisions of SSCs. In this review, we first address the morphological features, phenotypic characteristics, and the potential of SSCs, and then we focus on the recent advances in defining the key signaling molecules and crucial signaling pathways regulating self-renewal and differentiation of SSCs. The association of aberrant expression of signaling molecules and cascades with abnormal spermatogenesis and testicular cancer are also discussed. Finally, we point out potential future directions to pursue in research on signaling pathways of SSCs.


Asunto(s)
Diferenciación Celular , Transducción de Señal , Espermatogénesis , Espermatogonias/fisiología , Animales , Humanos , Masculino , Espermatogonias/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA