Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Endocrinol Metab ; 108(10): 2597-2603, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-36974363

RESUMEN

CONTEXT: Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) have the potential to improve native kidney function. OBJECTIVE: This work aimed to elucidate the possible protective effects of GLP-1 RAs on kidney graft function after successful kidney transplantation (KTX). METHODS: This retrospective cohort study included all KTX recipients (KTRs) at our facility with type 2 diabetes who were followed up from 1 month post-transplantation for 24 months or longer as of December 31, 2020. We investigated associations between the use of GLP-1 RAs and other antidiabetic medications (non-GLP-1 RAs) and the risk of sustained estimated glomerular filtration rate (eGFR) reduction (40% reduction compared with baseline for 4 months) for KTRs with type 2 diabetes. We calculated the propensity score of initiating GLP-1 RAs compared with that of initiating non-GLP-1 RAs as a function of baseline covariates using logistic regression. The inverse probability of the treatment-weighted odds ratio was estimated to control for baseline confounding variables. Sodium-glucose cotransporter 2 inhibitor use was a competing event. The primary outcome was sustained eGFR reduction of at least 40% from baseline for 4 months post-transplantation. RESULTS: Seventy-three patients were GLP-1 RA users and 73 were non-GLP-1 RA users. Six patients and 1 patient in the non-GLP-1 RA and GLP-1 RA groups had sustained eGFR reduction. GLP-1 RA use after KTX was associated with a lower risk of sustained eGFR reduction. CONCLUSION: GLP-1 RAs resulted in lower eGFR reduction compared with non-GLP-1 RAs and may contribute to better kidney graft survival after KTX.


Asunto(s)
Diabetes Mellitus Tipo 2 , Trasplante de Riñón , Insuficiencia Renal , Humanos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/cirugía , Diabetes Mellitus Tipo 2/complicaciones , Estudios Retrospectivos , Hipoglucemiantes/uso terapéutico , Péptido 1 Similar al Glucagón/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón/agonistas
2.
Front Endocrinol (Lausanne) ; 13: 941166, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35903276

RESUMEN

Human stem cell-derived organoid culture enables the in vitro analysis of the cellular function in three-dimensional aggregates mimicking native organs, and also provides a valuable source of specific cell types in the human body. We previously established organoid models of the hypothalamic-pituitary (HP) complex using human pluripotent stem cells. Although the models are suitable for investigating developmental and functional HP interactions, we consider that isolated pituitary cells are also useful for basic and translational research on the pituitary gland, such as stem cell biology and regenerative medicine. To develop a method for the purification of pituitary cells in HP organoids, we performed surface marker profiling of organoid cells derived from human induced pluripotent stem cells (iPSCs). Screening of 332 human cell surface markers and a subsequent immunohistochemical analysis identified epithelial cell adhesion molecule (EpCAM) as a surface marker of anterior pituitary cells, as well as their ectodermal precursors. EpCAM was not expressed on hypothalamic lineages; thus, anterior pituitary cells were successfully enriched by magnetic separation of EpCAM+ cells from iPSC-derived HP organoids. The enriched pituitary population contained functional corticotrophs and their progenitors; the former responded normally to a corticotropin-releasing hormone stimulus. Our findings would extend the applicability of organoid culture as a novel source of human anterior pituitary cells, including stem/progenitor cells and their endocrine descendants.


Asunto(s)
Células Madre Pluripotentes Inducidas , Hormonas Adenohipofisarias , Células Madre Pluripotentes , Biomarcadores/metabolismo , Molécula de Adhesión Celular Epitelial/metabolismo , Humanos , Organoides/metabolismo , Hipófisis/metabolismo , Hormonas Adenohipofisarias/metabolismo
3.
Sci Rep ; 11(1): 10729, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-34031474

RESUMEN

For pituitary regenerative medicine, the creation of a hypophyseal model in monkeys is necessary to conduct future preclinical studies; however, previous studies reported that hypophysectomy in monkeys is not always safe or satisfactory. This study aimed to create a hypophyseal dysfunction model in a cynomolgus monkey using a safer surgical technique and establish the protocol of pituitary hormone replacement therapy for this model. Surgical resection of the pituitary gland of a 7.8-year-old healthy adult cynomolgus male monkey weighing 5.45 kg was performed to create a hypophyseal dysfunction model for future regenerative studies. Endoscopic transoral transsphenoidal surgery was used to perform hypophysectomy under navigation support. These procedures were useful for confirming total removal of the pituitary gland without additional bone removal and preventing complications such as cerebrospinal fluid leakage. Total removal was confirmed by pathological examination and computed tomography. Hypopituitarism was verified with endocrinological examinations including stimulation tests. Postoperatively, the monkey's general condition of hypopituitarism was treated with hormone replacement therapy, resulting in long-term survival. The success of a minimally invasive and safe surgical method and long-term survival indicate the creation of a hypophyseal dysfunction model in a cynomolgus monkey; hence, this protocol can be employed in the future.


Asunto(s)
Hipofisectomía/efectos adversos , Hipopituitarismo/diagnóstico por imagen , Hipopituitarismo/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Terapia de Reemplazo de Hormonas , Humanos , Hipopituitarismo/etiología , Macaca fascicularis , Masculino , Procedimientos Quirúrgicos Mínimamente Invasivos , Medicina Regenerativa , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
4.
Cell Rep ; 30(1): 18-24.e5, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31914385

RESUMEN

The pituitary is a major hormone center that secretes systemic hormones responding to hypothalamus-derived-releasing hormones. Previously, we reported the independent pituitary induction and hypothalamic differentiation of human embryonic stem cells (ESCs). Here, a functional hypothalamic-pituitary unit is generated using human induced pluripotent stem (iPS) cells in vitro. The adrenocorticotropic hormone (ACTH) secretion capacity of the induced pituitary reached a comparable level to that of adult mouse pituitary because of the simultaneous maturation with hypothalamic neurons within the same aggregates. Corticotropin-releasing hormone (CRH) from the hypothalamic area regulates ACTH cells similarly to our hypothalamic-pituitary axis. Our induced hypothalamic-pituitary units respond to environmental hypoglycemic condition in vitro, which mimics a life-threatening situation in vivo, through the CRH-ACTH pathway, and succeed in increasing ACTH secretion. Thus, we generated powerful hybrid organoids by recapitulating hypothalamic-pituitary development, showing autonomous maturation on the basis of interactions between developing tissues.


Asunto(s)
Hipotálamo/fisiología , Células Madre Pluripotentes Inducidas/citología , Hipófisis/fisiología , Hormona Adrenocorticotrópica/metabolismo , Envejecimiento/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Corticotrofos/citología , Corticotrofos/ultraestructura , Humanos , Células Madre Pluripotentes Inducidas/ultraestructura , Ratones , Neuronas/citología , Organoides/citología
5.
Stem Cell Res ; 40: 101572, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31539858

RESUMEN

High differentiation efficiency is one of the most important factors in developing an in vitro model from pluripotent stem cells. In this report, we improved the handling technique applied to mouse-induced pluripotent stem (iPS) cells, resulting in better differentiation into hypothalamic vasopressin (AVP) neurons. We modified the culture procedure to make the maintenance of iPS cells in an undifferentiated state much easier. Three-dimensional floating culture was demonstrated to be effective for mouse iPS cells. We also improved the differentiation method with regards to embryology, resulting in a greater number of bigger colonies of AVP neurons differentiating from mouse iPS cells. Fgf8, which was not used in the original differentiation method, increased iPS differentiation into AVP neurons. These refinements will be useful as a valuable tool for the modeling of degenerative disease in AVP neurons in vitro using disease-specific iPS cells in future studies.


Asunto(s)
Diferenciación Celular , Línea Celular/citología , Hipotálamo/citología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/citología , Animales , Línea Celular/metabolismo , Células Cultivadas , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Hipotálamo/metabolismo , Células Madre Pluripotentes Inducidas/citología , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Vasopresinas/metabolismo
6.
Curr Protoc Neurosci ; 83(1): e48, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-30040229

RESUMEN

The anterior pituitary gland produces several hormones essential for regulation of the somatic endocrine system. Deficiency of these hormones can cause life-threatening diseases, including adrenal crisis. Pituitary tissue generated from human pluripotent stem cells is expected to provide better treatment than current hormone replacement therapy. During early mammalian development, the pituitary anlage (Rathke's pouch) develops from non-neural ectoderm adjacent to the developing ventral hypothalamus. The close interaction between these two tissues is crucial for Rathke's pouch development and involves several signaling molecules. Early exposure of human embryonic stem cells in 3D floating culture to sonic hedgehog and bone morphogenetic protein 4 promoted the cells' differentiation into oral ectoderm and, subsequently, hormone-producing cells such as corticotrophs (adrenocorticotropic hormone-producing cells). The differentiation approach described herein, which induces the formation of pituitary tissue in contact with hypothalamic neural tissue, mimics mammalian pituitary development. The differentiated corticotrophs are functional, responding normally to both release and feedback signals. © 2018 by John Wiley & Sons, Inc.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Embrionarias Humanas/patología , Hipófisis/patología , Células Madre Pluripotentes/patología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Desarrollo Embrionario/fisiología , Humanos
7.
Sci Rep ; 8(1): 3615, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29483626

RESUMEN

Arginine-vasopressin (AVP) neurons exist in the hypothalamus, a major region of the diencephalon, and play an essential role in water balance. Here, we established the differentiation method for AVP-secreting neurons from human embryonic stem cells (hESCs) by recapitulating in vitro the in vivo embryonic developmental processes of AVP neurons. At first, the differentiation efficiency was improved. That was achieved through the optimization of the culture condition for obtaining dorsal hypothalamic progenitors. Secondly, the induced AVP neurons were identified by immunohistochemistry and these neurons secreted AVP after potassium chloride stimulation. Additionally, other hypothalamic neuropeptides were also detected, such as oxytocin, corticotropin-releasing hormone, thyrotropin-releasing hormone, pro-opiomelanocortin, agouti-related peptide, orexin, and melanin-concentrating hormone. This is the first report describing the generation of secretory AVP neurons derived from hESCs. This method will be applicable to research using disease models and, potentially, for regenerative medicine of the hypothalamus.


Asunto(s)
Arginina Vasopresina/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteína Relacionada con Agouti/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Humanos , Hormonas Hipotalámicas/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Inmunohistoquímica , Melaninas/metabolismo , Neurofisinas/metabolismo , Orexinas/metabolismo , Oxitocina/metabolismo , Hormonas Hipofisarias/metabolismo , Precursores de Proteínas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Vasopresinas/metabolismo
8.
Methods Mol Biol ; 1597: 57-65, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28361310

RESUMEN

The adenohypophysis, which mainly consists of anterior pituitary, plays important roles for endocrine systems by secreting several hormones indispensable for maintaining homeostasis. During early mouse development, the pituitary primordium (called Rathke's pouch) develops from oral ectoderm adjacent to ventral hypothalamus by interaction between these two tissues. By using mouse embryonic stem cells (ESCs), we recapitulated this in vivo micro-environment of the pituitary development and demonstrated that Rathke's pouch-like structures were self-formed from three-dimensional (3D) floating culture. The mouse ESC-derived Rathke's pouch-like structures subsequently differentiated into hormone-producing cells such as corticotrophs and somatotrophs. We have modified this technique for human pluripotent stem cells and recently reported that pituitary placodes can also be generated from human ESCs through a similar process. Here, we describe a protocol for human ESC culture for in vitro generation of 3D pituitary tissue.


Asunto(s)
Hipófisis/citología , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Ectodermo/citología , Células Madre Embrionarias/citología , Humanos , Ratones , Adenohipófisis/citología , Células Madre Pluripotentes/citología
9.
Nat Commun ; 7: 10351, 2016 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-26762480

RESUMEN

Anterior pituitary is critical for endocrine systems. Its hormonal responses to positive and negative regulators are indispensable for homeostasis. For this reason, generating human anterior pituitary tissue that retains regulatory hormonal control in vitro is an important step for the development of cell transplantation therapy for pituitary diseases. Here we achieve this by recapitulating mouse pituitary development using human embryonic stem cells. We find that anterior pituitary self-forms in vitro following the co-induction of hypothalamic and oral ectoderm. The juxtaposition of these tissues facilitated the formation of pituitary placode, which subsequently differentiated into pituitary hormone-producing cells. They responded normally to both releasing and feedback signals. In addition, after transplantation into hypopituitary mice, the in vitro-generated corticotrophs rescued physical activity levels and survival of the hosts. Thus, we report a useful methodology for the production of regulator-responsive human pituitary tissue that may benefit future studies in regenerative medicine.


Asunto(s)
Corticotrofos/trasplante , Células Madre Embrionarias/fisiología , Hipopituitarismo/terapia , Adenohipófisis/embriología , Ingeniería de Tejidos/métodos , Hormona Adrenocorticotrópica/metabolismo , Animales , Técnicas de Cultivo de Célula , Desarrollo Embrionario , Hormona del Crecimiento/metabolismo , Humanos , Ratones , Adenohipófisis/metabolismo
10.
Curr Eye Res ; 41(4): 558-68, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-25880804

RESUMEN

PURPOSE: To establish a practical research tool for studying the pathogenesis of retinal ganglion cell (RGC) diseases, we optimized culture procedures to induce neurite outgrowth from three-dimensional self-organizing optic vesicles (3D-retinas) differentiated in vitro from mouse and human embryonic stem cells (ESCs). MATERIALS AND METHODS: The developing 3D-retinas isolated at various time points were placed on Matrigel-coated plates and cultured in media on the basis of the 3D-retinal culture or the retinal organotypic culture protocol. The number, length, and morphology of the neurites in each culture condition were compared. RESULTS: First, we confirmed that Venus-positive cells were double-labeled with a RGC marker, Brn3a, in the 3D-retina differentiated from Fstl4::Venus mouse ESCs, indicating specific RGC-subtype differentiation. Second, Venus-positive neurites grown from these RGC subsets were positive for beta-III tubulin and SMI312 by immunohistochemistry. Enhanced neurite outgrowth was observed in the B27-supplemented Neurobasal-A medium on Matrigel-coated plates from the optic vesicles isolated after 14 days of differentiation from mouse ESCs. For the differentiated RGCs from human ESCs, we obtained neurite extension of >4 mm by modifying Matrigel coating and the culture medium from the mouse RGC culture. CONCLUSION: We successfully optimized the culture conditions to enhance lengthy and high-frequency neurite outgrowth in mouse and human models. The procedure would be useful for not only developmental studies of RGCs, including maintenance and projection, but also clinical, pathological, and pharmacological studies of human RGC diseases.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Degeneración Nerviosa/terapia , Neuritas/patología , Proyección Neuronal/fisiología , Células Ganglionares de la Retina/patología , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Humanos , Imagenología Tridimensional , Inmunohistoquímica , Ratones , Ratones Noqueados , Degeneración Nerviosa/patología
11.
Stem Cell Res ; 15(2): 290-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26209816

RESUMEN

During embryonic development, oral ectoderm differentiates into the adenohypophysis, dental epithelia, salivary glands, and nasal pit. Few reports exist concerning the induction of oral ectoderm from embryonic stem (ES) cells. Generally, any lot differences in fetal bovine serum (FBS) and serum replacer may affect the induction of ES cell-differentiation. Using a previously established culture strategy for differentiation, the proportion of cell aggregates containing Pitx1+ oral ectoderm varied widely between 9-36% when several different lots of FBS or serum replacer were used. We therefore tried to enhance the differentiation method. We found that bone morphogenetic protein (BMP) 4 and fibroblast growth factor (FGF) treatments improved oral ectoderm induction. Such treatment also improved the differentiation of oral ectoderm into the adenohypophysis. Furthermore, increased BMP4 treatment induced dental epithelium and mesenchyme. Such differentiation suggests that the Pitx1+ layer displays similar properties to oral ectoderm, as found in vivo. Differentiation of ES cells into oral ectoderm using different lots of FBS and serum replacer increased 78-90% after treatment with BMP4 and FGF. In summary, we have established a robust strategy for the induction of oral ectoderm differentiation from mouse ES cells.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Animales , Ectodermo/citología , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factor de Transcripción Pit-1/genética , Factor de Transcripción Pit-1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína del Homeodomínio PITX2
12.
Nat Commun ; 6: 6286, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25695148

RESUMEN

In the developing neural retina (NR), multipotent stem cells within the ciliary margin (CM) contribute to de novo retinal tissue growth. We recently reported the ability of human embryonic stem cells (hESCs) to self-organize stratified NR using a three-dimensional culture technique. Here we report the emergence of CM-like stem cell niches within human retinal tissue. First, we developed a culture method for selective NR differentiation by timed BMP4 treatment. We then found that inhibiting GSK3 and FGFR induced the transition from NR tissue to retinal pigment epithelium (RPE), and that removing this inhibition facilitated the reversion of this RPE-like tissue back to the NR fate. This step-wise induction-reversal method generated tissue aggregates with RPE at the margin of central-peripherally polarized NR. We demonstrate that the NR-RPE boundary tissue further self-organizes a niche for CM stem cells that functions to expand the NR peripherally by de novo progenitor generation.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Embrionarias/citología , Retina/embriología , Nicho de Células Madre/fisiología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Linaje de la Célula , Femenino , Citometría de Flujo , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos ICR , Células Madre Multipotentes/citología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Retina/citología , Epitelio Pigmentado de la Retina/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...