lung progenitors
Recently Published Documents


TOTAL DOCUMENTS

26
(FIVE YEARS 12)

H-INDEX

5
(FIVE YEARS 2)

Biomaterials ◽  
2021 ◽  
pp. 121031
Author(s):  
Satoshi Ikeo ◽  
Yuki Yamamoto ◽  
Kazuhiro Ikeda ◽  
Naoyuki Sone ◽  
Yohei Korogi ◽  
...  

2021 ◽  
Author(s):  
Renee F.C. Hein ◽  
Joshua H. Wu ◽  
Yu-Hwai Tsai ◽  
Angeline Wu ◽  
Alyssa J. Miller ◽  
...  

SUMMARYMammalian respiratory system development is regulated by complex reciprocal signaling events that take place between epithelial cells and the surrounding mesenchymal cells; however, mesenchymal heterogeneity and function in the developing human lung is poorly understood. We interrogated single cell RNA sequencing data from multiple human lung specimens and identified a mesenchymal cell population present during development that is highly enriched for expression of the WNT agonist R-SPONDIN2 (RSPO2), and we found that adjacent epithelial bud tip progenitors are enriched for the RSPO2 receptor LGR5. By carrying out functional experiments using organoid models, lung explant cultures, and FACS-isolated RSPO2+ mesenchyme, we show that RSPO2 is a critical niche cue that potentiates WNT signaling in human lung progenitors to maintain their multipotency.


2021 ◽  
Vol 16 (4) ◽  
pp. 1802-1829
Author(s):  
Ana Luisa Rodrigues Toste de Carvalho ◽  
Hsiao-Yun Liu ◽  
Ya-Wen Chen ◽  
Matteo Porotto ◽  
Anne Moscona ◽  
...  

2021 ◽  
Author(s):  
Chaido Ori ◽  
Meshal Ansari ◽  
Ilias Angelidis ◽  
Fabian J. Theis ◽  
Herbert B. Schiller ◽  
...  

ABSTRACTUnderstanding the development of human respiratory tissues is crucial for modeling and treating lung disorders. The molecular details for the specification of lung progenitors from human pluripotent stem cells (hPSCs) are unclear. Here, we use single cell RNA-sequencing with high temporal resolution along an optimized differentiation protocol to determine the transcriptional hierarchy of lung specification from human hPSCs and map out the underlying single cell trajectories. We show that Sonic hedgehog, TGF-β and Notch activation are required in an ISL1/NKX2-1 trajectory that gives rise to lung progenitors during the foregut endoderm stage. Induction of HHEX marks an alternative trajectory at the early definitive endoderm stage, which precedes the lung trajectory and generates a major hepatoblast population. Moreover, neither KDR+ nor mesendoderm progenitors are apparent intermediate states of lung and hepatic lineages. Our hierarchical multistep model predicts mechanisms leading to lung organogenesis, and creates a basis for studying early human lung development, as well as hPSC based disease and drug research.Abstract Figure


Biomaterials ◽  
2020 ◽  
Vol 254 ◽  
pp. 120128 ◽  
Author(s):  
John P. Soleas ◽  
Elisa D'Arcangelo ◽  
Linwen Huang ◽  
Golnaz Karoubi ◽  
Maria Cristina Nostro ◽  
...  
Keyword(s):  

2020 ◽  
Vol 26 (4) ◽  
pp. 593-608.e8 ◽  
Author(s):  
Killian Hurley ◽  
Jun Ding ◽  
Carlos Villacorta-Martin ◽  
Michael J. Herriges ◽  
Anjali Jacob ◽  
...  

2019 ◽  
Author(s):  
Killian Hurley ◽  
Jun Ding ◽  
Carlos Villacorta-Martin ◽  
Michael J. Herriges ◽  
Anjali Jacob ◽  
...  

AbstractAlveolar epithelial type 2 cells (AEC2s) are the facultative progenitors responsible for maintaining lung alveoli throughout life, yet are difficult to access from patients for biomedical research or lung regeneration applications. Here we engineer AEC2s from human induced pluripotent stem cells (iPSCs) in vitro and use single cell RNA sequencing (scRNA-seq) to profile the detailed kinetics of their differentiation over time. We focus on both the desired target cells as well as those that appear to diverge to alternative endodermal fates. By combining scRNA-seq with lentiviral barcoding to trace differentiating clones, we reveal the bifurcating cell fate trajectories followed as primordial lung progenitors differentiate into mature AEC2s. We define the global transcriptomic signatures of primary developing human AEC2s from fetal through adult stages in order to identify the subset of in vitro differentiating cells that appear to recapitulate the path of in vivo development. In addition, we develop computational methods based on Continuous State Hidden Markov Models (CSHMM) to identify the precise timing and type of signals, such as over-exuberant Wnt responses, that induce some early multipotent NKX2-1+ progenitors to lose lung fate as they clonally diverge into a variety of non-lung endodermal lineages. Finally, we find that this initial developmental plasticity is regulatable via Wnt modulation, and subsides over time, ultimately resulting in iPSC-derived AEC2s that exhibit a stable phenotype and nearly limitless self-renewal capacity in vitro. Our methods and computational approaches can be widely applied to study and control directed differentiation, producing an inexhaustible supply of mature lineages, exemplified here by the generation of AEC2s.


Sign in / Sign up

Export Citation Format

Share Document