Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-07
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • Our study made use of single cell transcriptional

    2018-11-02

    Our study made use of single-cell transcriptional profiling to define the heterogeneity of hormone-producing chemokine receptor derived from hPSCs. The technology is a powerful tool that allowed us to determine the percentage of various hormone-producing cells and shifts in this distribution upon treatment with patterning factors or comparing different time points in vitro. However, one of the challenges of single-cell profiling is the largely binary readout of the expression data (McDavid et al., 2013). The HESX1/NEUROD1 validation experiment exemplifies this drawback, as the single-cell RT-PCR was not able to capture the decrease in HESX1 expression over time as observed by immunocytochemistry. Therefore, our binary approach of identifying expressed genes leads to the possibility that lowly expressed genes may be read as positive despite their expression levels not being high enough to achieve meaningful translation into protein. For example, we observed some instances of “inappropriate” expression patterns such as the co-expression of SIX1 with PAX6 and PAX3 (though at different expression levels), which would indicate a mixed regional cranial placode identity. Similarly, the presence of putative plurihormonal cells was based purely on the expression of multiple hormonal transcripts. The presence of plurihormonal cells in the pituitary gland has been reported previously (Nunez et al., 2003; Villalobos et al., 2004) and is a well-known feature in pituitary adenomas (Scheithauer et al., 1986). However, it will be important to further assess in hPSC-derived pituitary cells whether transcripts for multiple hormones indeed result in the expression and secretion of multiple hormones from individual cells. To resolve some of the limitations of the single-cell gene-expression study, it will be important in the future to obtain appropriate positive and negative controls in order to define tissue-relevant thresholds of expression for each transcript. Unfortunately, developmentally matched human tissues are not readily available. In the present study we used immunofluorescence analysis as an independent method to quantify bona fide hormone-producing cells. Due to the lower sensitivity of this assay compared with qRT-PCR analysis, the percentages of hormone-positive cells were found to be lower but with the same relative proportions of the various hormone-producing cells. We demonstrate that exposure to FGF8 and BMP2 can bias the dorsal-ventral composition of hormone-producing cells from ACTH+ cells to FSH+ or LH+ cells. It should be noted that in vivo, cell types expressing different hormones are not necessarily located in specific areas but are scattered throughout the mature gland (Ericson et al., 1998; Olson et al., 2006). In addition, some reports argue for a less important role of extrinsic signaling compared with intrinsic factors in pituitary cell specification (Davis et al., 2011). Our human in vitro system provides a valuable tool to answer such questions. The current differentiation protocols only achieve partial enrichment of specific hormonal lineages. Therefore, lineage selection via cell sorting or the development of more sophisticated patterning strategies will be required to obtain purified populations such as GH+ cells suitable for treating patients with selective GH deficiency (Bianchi et al., 2008). Our cell-surface epitope screen presents candidate markers that may be suitable for isolating specific hormone lineages. One cell type lacking in the current in vitro culture system are TSH+ cells. Studies in sheep suggest that proper hypothalamic input is required for the development of thyrotrophs (Szarek et al., 2008). Therefore, it is conceivable that hypothalamic lineage cells may still be required for generating TSH+ fates. Alternatively, TSH+ cells may simply require further optimization of our defined induction conditions or longer in vitro differentiation periods. The in vivo studies show clear evidence of survival and hormone release up to 7 weeks after transplantation. Whether the lower percentage of hormone-producing cells in vivo is due to limited cell survival or increased proliferation of immature pituitary progenitors remains to be determined. Future studies also need to define the optimal transplantation time point for in vivo yield and hormone subtype and demonstrate the long-term survival of hPSC-derived pituitary cells. One key question is whether the functional integration of human cells will eventually require orthotopic placement of the graft into the pituitary gland or hypothalamic region. Preliminary experiments with orthotopic (median eminence) placement of the cells into lesioned animals resulted in low cell survival, suggesting the need for further optimization of such a transplantation paradigm. Ectopic placement in accessible locations does offer advantages from a translational perspective, namely the ability to access and remove the graft, should adverse side effects develop. However, hypothalamic release factors are the main triggers for hormonal release in the anterior pituitary gland (Smith and Vale, 2006). These release hormones have a short half-life and do not persist in the circulation. They are released into the hypophyseal portal venous plexus, which ensures immediate local delivery to the gland. Thus it is likely that the ectopically placed hPSC-derived grafts did not gain significant exposure to hypothalamic input. Our data therefore suggest a degree of autonomous hormonal release by the grafted cells, although a more comprehensive assessment of integration into homeostatic, endocrine mechanisms will be necessary. Based on existing literature on grafting primary cells (Maxwell et al., 1998; Naik et al., 1997), it is likely that proper regulation of at least some of the pituitary hormones will require orthotopic placement. Direct access to the skull base and pituitary gland in the mouse requires aggressive surgical procedures, such as transaural approaches or parapharyngeal neck dissection, complicating such studies. This is in marked contrast to human patients in whom surgical access to the pituitary gland can be safely performed through minimally invasive endoscopic transnasal routes (Tabar, 2011).