Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Dec 18;15(1):480.
doi: 10.1186/s13287-024-04080-w.

ZO-1 boosts the in vitro self-renewal of pre-haematopoietic stem cells from OCT4-reprogrammed human hair follicle mesenchymal stem cells through cytoskeleton remodeling

Affiliations

ZO-1 boosts the in vitro self-renewal of pre-haematopoietic stem cells from OCT4-reprogrammed human hair follicle mesenchymal stem cells through cytoskeleton remodeling

Yingchun Ruan et al. Stem Cell Res Ther. .

Erratum in

Abstract

Background: The challenge of expanding haematopoietic stem/progenitor cells (HSPCs) in vitro has limited their clinical application. Human hair follicle mesenchymal stem cells (hHFMSCs) can be reprogrammed to generate intermediate stem cells by transducing OCT4 (hHFMSCsOCT4) and pre-inducing with FLT3LG/SCF, and differentiated into erythrocytes. These intermediate cells exhibit gene expression patterns similar to pre-HSCs, making them promising for artificial haematopoiesis. However, further investigation is required to elucidate the in vitro proliferation ability and mechanism underlying the self-renewal of pre-HSCs derived from hHFMSCs.

Methods: hHFMSCsOCT4 were pre-treated with FLT3LG and SCF cytokines, followed by characterization and isolation of the floating cell subsets for erythroid differentiation through stimulation with hematopoietic cytokines and nutritional factors. Cell adhesion was assessed through disassociation and adhesion assays. OCT4 expression levels were measured using immunofluorescence staining, RT-qPCR, and Western blotting. RNA sequencing and Gene Ontology (GO) enrichment analysis were then conducted to identify proliferation-related biological processes. Proliferative capacity was evaluated through CCK-8, colony formation assays, Ki67 index, and cell cycle analysis. Cytoskeleton was observed through Wright‒Giemsa, Coomassie brilliant blue, and phalloidin staining. Expression of adherens junction (AJ) core members was confirmed through RT‒qPCR, Western blotting, and immunofluorescence staining before and after ZO-1 knockdown. A regulatory network was constructed to determine relationships among cytoskeleton, proliferation, and the AJ pathway. Student's t tests (GraphPad Prism 8.0.2) were used for group comparisons. The results were considered significant at P < 0.05.

Results: Pre-treatment of hHFMSCsOCT4 with FLT3LG and SCF leads to the emergence of floating cell subsets exhibiting small, globoid morphology, suspended above adherent cells, forming colonies, and displaying minimal expression of CD45. Excessive OCT4 expression weakens adhesion in floating hHFMSCsOCT4. Floating cells moderately enhanced proliferation and undergo cytoskeleton remodelling, with increased contraction and aggregation of F-actin near the nucleus. The upregulation of ZO-1 could impact the expressions of F-actin, E-cadherin, and β-catenin genes, as well as the nuclear positioning of β-catenin, leading to variations in the cytoskeleton and cell cycle. Finally, a regulatory network revealed that the AJ pathway cored with ZO-1 critically bridges cytoskeletal remodelling and haematopoiesis-related proliferation in a β-catenin-dependent manner.

Conclusions: ZO-1 improved the self-renewal of pre-HSCs from OCT4-overexpressing hHFMSCs by remodeling the cytoskeleton via the ZO-1-regulated AJ pathway, suggesting floating hHFMSCsOCT4 as the promising seed cells for artificial hematopoiesis.

Keywords: Adherens junction; Human hair follicle mesenchymal stem cells; Pre-haematopoietic stem cells; Reprogramming; Self-renewal.

PubMed Disclaimer

Conflict of interest statement

Declarations. Ethics approval and consent to participate: Informed consent was obtained from the donors to isolated hHFMSCs for research use. All experiments were approved by the Medical Ethics Committee of Qingdao Municipal Hospital. Title of the approved project: OCT4 facilitates the transdifferentiation of human hair follicle stem cells into erythrocytes by remodeling the cytoskeleton. Name of the institutional approval committee or unit: Medical Ethics Committee/IRB of Qingdao Municipal Hospital. Approval number: 2024-KY-077, Date of approval: Oct 16, 2024. Consent for publication: Informed consent was obtained from the donors to isolated hHFMSCs for research use. All authors have reviewed the manuscript and approved the publication. Competing interests: The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
FLT3LG and SCF caused floating cells to appear from hHFMSCsOCT4. (a) Schema showing the transformation of floating cells derived from hHFMSCsOCT4 into erythroblasts. (b) Cell morphology changes and floating cells observed under immunofluorescence microscope, with a floating cell indicated by a yellow arrow and a floating cell colony outlined by a dashed circle. (c) Phase contrast microscope used to observe floating cells’ morphology. Floating cell indicated by yellow arrow. (d) Collecting and re-cultivating suspended cells. (e and f). Floating cell colonies were selected to identify CD45 expression, with green indicating EGFR, red indicating PE-CD45, and blue indicating Hoechst. Peripheral blood cells served as a positive control
Fig. 2
Fig. 2
Floating hHFMSCsOCT4 differentiated into erythrocytes. (a) Erythroblast differentiation and cell morphology change in response to hematopoietic cytokines. Expansion D7 refers to erythroid cells expanded for 7 days in erythroid cell medium. (b) Erythroblast growth and maturation in erythroid expansion medium with nutrients. Green arrow: promyelocyte and mesmyelocyte. Orange arrow: metarubricyte. Red arrows: enucleated erythrocyte. (c) Erythrocytes were enucleated in a culture medium without cytokines and the enucleation efficiency was measured
Fig. 3
Fig. 3
The difference in OCT4 overexpression between adherent hHFMSCsOCT4 and floating hHFMSCsOCT4. (a) IF analysis of OCT4 expression and localization in hHFMSCs, adherent hHFMSCsOCT4, and floating hHFMSCsOCT4. NC, negative control. Red represents OCT4, blue represents DAPI, and green represents EGFP, (n = 3). (b) and (c) Histograms were generated to validate the expression of OCT4 by RT‒qPCR (n = 3) and (d) Western blots (n = 1), respectively. Full-length blots/gels are presented in Supplementary Figure S3. **P˂0.01, ***P˂0.001, ****P˂0.0001
Fig. 4
Fig. 4
The difference in self-renewal and proliferative ability between the adherent hHFMSCsOCT4 and floating hHFMSCsOCT4. (a) Cell proliferation curve of hHFMSCs, adherent hHFMSCsOCT4 and floating hHFMSCsOCT4 obtained from the CCK-8 assay (n = 3). (b) Colony formation assay of hHFMSCs, adherent hHFMSCsOCT4, and floating hHFMSCsOCT4; the enlarged views show the difference between the three cell clones (n = 3). The histogram is the rate of colony formation for each cell. (c) ICC of proliferation-associated protein Ki67 expression in three cell groups, and Ki67 protein was localized in the nucleus (n = 3). *P˂0.05, **P˂0.01, ****P˂0.0001. (d) Discrete cutoffs for distinct cell cycle phases are visually selected to calculate the percentages of cells in G1, S, and G2/M phases using R software
Fig. 5
Fig. 5
Remodelling of the cytoskeleton in floating hHFMSCsOCT4. (a) The cytoplasm and nucleus were stained with Wright‒Giemsa stain in three groups of cells (n = 3). (b) Cytoskeletal microfilaments in the three groups were stained with Coomassie brilliant blue (n = 3). (c) F-actin morphology and distribution were detected by phalloidin staining (n = 3). These images were all taken from randomly selected fields. *P˂0.05, **P˂0.01
Fig. 6
Fig. 6
Expression of AJ core genes and target genes in floating hHFMSCsOCT4. (a) RT‒qPCR analysis of the expression of ZO-1, ACTN2, E-cadherin, β-catenin, and Cyclin D1 (n = 3). (b) IF staining images showing the location of the β-catenin protein in hHFMSCs, adherent hHFMSCsOCT4, and floating hHFMSCsOCT4. (c) Fluorescence intensity of the merged colour in the nucleus was measured in three groups using ImageJ. *P˂0.05, **P˂0.01, ***P˂0.001
Fig. 7
Fig. 7
Changes in AJ gene expression and F-actin arrangement in floating hHFMSCsOCT4 after ZO-1 knockdown. (a) RT‒qPCR and Western blot showing the expression of ZO-1 after RNAi (n = 2). Full-length blots/gels are presented in Supplementary Figure S4. (b) RT‒qPCR and detection of the mRNA expression of ACTN2, E-cadherin, β-catenin, and Cyclin D1 (n = 3). The error bars represent the standard deviations of measurements in three separate sample runs. (c) Western blots showing the protein expression levels of β-catenin (n = 1). Full-length blots/gels are presented in Supplementary Figure S5, n = 1. (d) Phalloidin staining showing F-actin morphology and distribution (n = 3). (e and f) Fluorescence microscopy (Olympus) captured DAPI-stained cell images, and CellProfiler software detected nuclei and calculated their integrated intensity. R software then used visually determined cutoffs to calculate the percentages of cells in G1, S, and G2/M phases. NC, negative control. si-ZO-1, ZO-1-knockdown. *P˂0.05, **P˂0.01, ***P˂0.001, ****P˂0.0001
Fig. 8
Fig. 8
Network analysis. A gene network regulates haematopoiesis-related proliferation of floating hHFMSCsOCT4. The solid line shows gene relationships within the group, while the dashed line shows relationships between genes in different groups

Similar articles

Cited by

References

    1. C D. Blood stem cells produced in vast quantities in the lab. Nature. 2019;570(7759):17–8. - PubMed
    1. Sakurai MIK, Ito R, Wilkinson AC, Kimura T, Mizutani E, Nishikii H, Sudo K, Becker HJ, Takemoto H, Sano T, Kataoka K, Takahashi S, Nakamura Y, Kent DG, Iwama A, Chiba S, Okamoto S, Nakauchi H, Yamazaki S. Chemically defined cytokine-free expansion of human haematopoietic stem cells. Nature. 2023;615(7950):127–33. - PubMed
    1. Boisset JCCT, Klaus A, Papazian N, Onderwater J, Mommaas-Kienhuis M, Cupedo T, Robin C. Progressive maturation toward hematopoietic stem cells in the mouse embryo aorta. Blood. 2015;125(3):465–9. - PMC - PubMed
    1. Li CCZG, Du J, Liu D, Li Z, Ni Y, Zhou J, Li Y, Hou S, Zheng X, Lan Y, Liu B, He A. Pre-configuring chromatin architecture with histone modifications guides hematopoietic stem cell formation in mouse embryos. Nat Commun. 2022;13(1):346. - PMC - PubMed
    1. Wang Y, Liu J, Tan X, Li G, Gao Y, Liu X, Zhang L, Li Y. Induced pluripotent stem cells from human hair follicle mesenchymal stem cells. Stem Cell Rev Rep. 2013;9(4):451–60. - PMC - PubMed

MeSH terms

Substances