Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Sci Rep ; 12(1): 17381, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36253431

ABSTRACT

Familial neurohypophyseal diabetes insipidus (FNDI) is a degenerative disease of vasopressin (AVP) neurons. Studies in mouse in vivo models indicate that accumulation of mutant AVP prehormone is associated with FNDI pathology. However, studying human FNDI pathology in vivo is technically challenging. Therefore, an in vitro human model needs to be developed. When exogenous signals are minimized in the early phase of differentiation in vitro, mouse embryonic stem cells (ESCs)/induced pluripotent stem cells (iPSCs) differentiate into AVP neurons, whereas human ESCs/iPSCs die. Human ESCs/iPSCs are generally more similar to mouse epiblast stem cells (mEpiSCs) compared to mouse ESCs. In this study, we converted human FNDI-specific iPSCs by the naive conversion kit. Although the conversion was partial, we found improved cell survival under minimal exogenous signals and differentiation into rostral hypothalamic organoids. Overall, this method provides a simple and straightforward differentiation direction, which may improve the efficiency of hypothalamic differentiation.


Subject(s)
Diabetes Insipidus, Neurogenic , Induced Pluripotent Stem Cells , Animals , Cell Differentiation , Humans , Hypothalamus/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Neurons/metabolism , Vasopressins/metabolism
2.
Cell Rep ; 30(1): 18-24.e5, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914385

ABSTRACT

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.


Subject(s)
Hypothalamus/physiology , Induced Pluripotent Stem Cells/cytology , Pituitary Gland/physiology , Adrenocorticotropic Hormone/metabolism , Aging/physiology , Animals , Cell Differentiation , Cells, Cultured , Corticotrophs/cytology , Corticotrophs/ultrastructure , Humans , Induced Pluripotent Stem Cells/ultrastructure , Mice , Neurons/cytology , Organoids/cytology
3.
Stem Cell Res ; 40: 101572, 2019 10.
Article in English | MEDLINE | ID: mdl-31539858

ABSTRACT

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.


Subject(s)
Cell Differentiation , Cell Line/cytology , Hypothalamus/cytology , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology , Animals , Cell Line/metabolism , Cells, Cultured , Fibroblast Growth Factor 8/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Hypothalamus/metabolism , Induced Pluripotent Stem Cells/cytology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Vasopressins/metabolism
4.
Endocrinology ; 160(7): 1701-1718, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31135891

ABSTRACT

Tanycytes have recently been accepted as neural stem/progenitor cells in the postnatal hypothalamus. Persistent retina and anterior neural fold homeobox (Rax) expression is characteristic of tanycytes in contrast to its transient expression of whole hypothalamic precursors. In this study, we found that Rax+ residual cells in the maturation phase of hypothalamic differentiation in mouse embryonic stem cell (mESC) cultures had similar characteristics to ventral tanycytes. They expressed typical neural stem/progenitor cell markers, including Sox2, vimentin, and nestin, and differentiated into mature neurons and glial cells. Quantitative RT-PCR analysis showed that Rax+ residual cells expressed Fgf-10, Fgf-18, and Lhx2, which are expressed by ventral tanycytes. They highly expressed tanycyte-specific genes Dio2 and Gpr50 compared with Rax+ early hypothalamic progenitor cells. Therefore, Rax+ residual cells in the maturation phase of hypothalamic differentiation were considered to be more differentiated and similar to late progenitor cells and tanycytes. They self-renewed and formed neurospheres when cultured with exogenous FGF-2. Additionally, these Rax+ neurospheres differentiated into three neuronal lineages (neurons, astrocytes, and oligodendrocytes), including neuropeptide Y+ neuron, that are reported to be differentiated from ventral tanycytes toward the arcuate nuclei. Thus, Rax+ residual cells were multipotent neural stem/progenitor cells. Rax+ neurospheres were stably passaged and retained high Sox2 expression even after multiple passages. These results suggest the successful induction of Rax+ tanycyte-like cells from mESCs [induced tanycyte-like (iTan) cells]. These hypothalamic neural stem/progenitor cells may have potential in regenerative medicine and as a research tool.


Subject(s)
Cell Lineage/physiology , Embryonic Stem Cells/metabolism , Ependymoglial Cells/metabolism , Hypothalamus/metabolism , Neural Stem Cells/metabolism , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Ependymoglial Cells/cytology , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factors/metabolism , Hypothalamus/cytology , LIM-Homeodomain Proteins/metabolism , Mice , Neural Stem Cells/cytology , Transcription Factors/metabolism
5.
Sci Rep ; 8(1): 3615, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483626

ABSTRACT

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.


Subject(s)
Arginine Vasopressin/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Agouti-Related Protein/metabolism , Corticotropin-Releasing Hormone/metabolism , Humans , Hypothalamic Hormones/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Immunohistochemistry , Melanins/metabolism , Neurophysins/metabolism , Orexins/metabolism , Oxytocin/metabolism , Pituitary Hormones/metabolism , Protein Precursors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Vasopressins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL