Ex vivo Expansion Potential of Murine Hematopoietic Stem Cells: A Rare Property Only Partially Predicted by Phenotype

  1. Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medical, Lund University, 221 84 Lund, Sweden

Editors

  • Reviewing Editor
    Dominique Bonnet
    The Francis Crick Institute, London, United Kingdom
  • Senior Editor
    Didier Stainier
    Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany

Reviewer #1 (Public Review):

In 2019, Wilkinson and colleagues (PMID: 31142833) managed to break the veil in a 20-year open question on how to properly culture and expand Hematopoietic Stem Cells (HSCs). Although this study is revolutionizing the HSC biology field, several questions regarding the mechanisms of expansion remain open. Leveraging on this gap, Zhang et al.; embarked on a much-needed investigation regarding HSC self-renewal in this particular culturing setting.

The authors firstly tacked the known caveat that some HSC membrane markers are altered during in vitro cultures by functionally establishing EPCR (CD201) as a reliable and stable HSC marker (Figure 1), demonstrating that this compartment is also responsible for long-term hematopoietic reconstitution (Figure 3). Next in Figure 2, the authors performed single-cell omics to shed light on the potential mechanisms involved in HSC maintenance, and interestingly it was shown that several hematopoietic populations like monocytes and neutrophils are also present in this culture conditions, which has not been reported. The study goes on to functionally characterize these cultured HSCs (cHSC). The authors elegantly demonstrate using state-of-the-art barcoding strategies that these culturing conditions provoke heterogeneity in the expanding HSC pool (Figure 4). In the last experiment (Figure 5), it was demonstrated that cHSC not only retain their high EPCR expression levels but upon transplantation, these cells remain more quiescent than freshly-isolated controls.

Taken together, this study independently validates that the proposed culturing system works and provides new insights into the mechanisms whereby HSC expansion takes place.

Most of the conclusions of this study are well supported by the present manuscript, some aspects regarding experimental design and especially the data analysis should be clarified and possibly extended.

1. The first major point regards the single-cell (sc) omics performed on whole cultured cells (Figure 2):
a. The authors claim that both RNA and ATAC were performed and indeed some ATAC-seq data is shown in Figure 2B, but this collected data seems to be highly underused.
b. It's not entirely clear to this reviewer the nature of the so-called "HSC signatures"(SF2C) and why exactly these genes were selected. There are genes such as Mpl and Angpt1 which are used for Mk-biased HSCs. Maybe relying on other HSC molecular signatures (PMID: 12228721, for example) would not only bring this study more into the current field context but would also have a more favorable analysis outcome. Moreover reclustering based on a different signature can also clarify the emergence of relevant HSC clusters.
c. The authors took the hard road to perform experiments with the elegant HSC-specific Fgd5-reporter, and they claim in lines 170-171 that it "failed to clearly demarcate in our single-cell multimodal data". This seems like a rather vague statement and leads to the idea that the scRNA-seq experiment is not reliable. It would be interesting to show a UMAP with this gene expression regardless and also potentially some other HSC markers.

2. During the discussion and in Figure 4, the authors ponder and demonstrate that this culturing system can provoke divert HSC close expansion, having also functional consequences. This a known caveat from the original system, but in more recent publications from the original group (PMID: 36809781 and PMID: 37385251) small alterations into the protocol seem to alleviate clone selection. It's intriguing why the authors have not included these parameters at least in some experiments to show reproducibility or why these studies are not mentioned during the discussion section.

3. In this reviewer's opinion, the finding that transplanted cHSC are more quiescent than freshly isolated controls is the most remarkable aspect of this manuscript. There is a point of concern and an intriguing thought that sprouts from this experiment. It is empirical that for this experiment the same HSC dose is transplanted between both groups. This however is technically difficult since the membrane markers from both groups are different. Although after 8 weeks chimerism levels seem to be the same (SF5D) for both groups, it would strengthen the evidence if the author could demonstrate that the same number of HSCs were transplanted in both groups, likely by limiting dose experiments. Finally, it's interesting that even though EE100 cells underwent multiple replication rounds (adding to their replicative aging), these cells remained more quiescent once they were in an in vivo setting. Since the last author of this manuscript has also expertise in HSC aging, it would be interesting to explore whether these cells have "aged" during the expansion process by assessing whether they display an aged phenotype (myeloid-skewed output in serial transplantations and/or assisting their transcriptional age).

Reviewer #2 (Public Review):

Summary:
In this study, Zhang and colleagues characterise the behaviour of mouse hematopoietic stem cells when cultured in PVA conditions, a recently published method for HSC expansion (Wilkinson et al., Nature, 2019), using multiome analysis (scRNA-seq and scATACseq in the same single cell) and extensive transplantation experiments. The latter are performed in several settings including barcoding and avoiding recipient conditioning. Collectively the authors identify several interesting properties of these cultures namely: 1) only very few cells within these cultures have long-term repopulation capacity, many others, however, have progenitor properties that can rescue mice from lethal myeloablation; 2) single-cell characterisation by combined scRNAseq and scATACseq is not sufficient to identify cells with repopulation capacity; 3) expanded HSCs can be engrafted in unconditioned host and return to quiescence.
The authors also confirm previous studies that EPCRhigh HSCs have better reconstitution capability than EPCRlow HSCs when transplanted.

Strengths:
The major strength of this manuscript is that it describes how functional HSCs are expanded in PVA cultures to a deeper extent than what has been done in the original publication. The authors are also mindful of considering the complexities of interpreting transplantation data. As these PVA cultures become more widely used by the HSC community, this manuscript is valuable as it provides a better understanding of the model and its limitations.

Novelty aspects include:
• The authors determined that small numbers of expanded HSCs enable transplantation into non-conditioned syngeneic recipients.
• This is to my knowledge the first report characterising the output of PVA cultures by multiome. This could be a very useful resource for the field.
• They are also the first to my knowledge to use barcoding to quantify HSC repopulation capacity at the clonal level after PVA culture.
• It is also useful to report that HSCs isolated from fetal livers do expand less than their adult counterparts in these PVA cultures.

Weaknesses:
• The analysis of the multiome experiment is limited. The authors do not discuss what cell types, other than functional or phenotypic HSCs are present in these cultures (are they mostly progenitors or bona fide mature cells?) and no quantifications are provided.
• Barcoding experiments are technically elegant but do not bring particularly novel insights.
• The number of mice analysed in certain experiments is fairly low (Figures 1 and 5).
• The manuscript remains largely descriptive. While the data can be used to make useful recommendations to future users working with PVA cultures and in general with HSCs, those recommendations could be more clearly spelled out in the discussion.
• The authors should also provide a discussion of the other publications that have used these methods to date.

Overall, the authors succeeded in providing a useful set of experiments to better interpret what type of HSCs are expanded in PVA cultures. More in-depth mining of their bioinformatic data (by the authors or other groups) is likely to highlight other interesting/relevant aspects of HSC biology in relation to this expansion methodology.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation