Hematopoietic Transfer of the Anti-Cancer and Lifespan-Extending Capabilities of A Genetically Engineered Blood System

  1. The PhD Program for Neural Regenerative Medicine, Taipei Medical University, Taipei 115, Taiwan
  2. Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan
  3. Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan
  4. Department of Nursing, Chang Gung University of Science and Technology, Taoyuan City 333, Taiwan
  5. Agricultural Biotechnology Research Center, Academia Sinica, Nankang, Taipei 115, Taiwan
  6. Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 115, Taiwan
  7. Genomics Research Center, Academia Sinica, Nankang, Taipei 115, Taiwan

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Yelena Ginzburg
    Icahn School of Medicine at Mount Sinai, New York, United States of America
  • Senior Editor
    Mone Zaidi
    Icahn School of Medicine at Mount Sinai, New York, United States of America

Reviewer #1 (Public Review):

The authors Wang et al. present a study of a mouse model K74R that they claim can extend the life span of mice, and also has some anti-cancer properties. Importantly, this mechanism seems to be mediated by the hematopoietic system, and protective effects can be transferred with bone marrow transplantation.

The authors need to be more specific in the title and abstract as to what is actually novel in this manuscript (a single tumor model), and what relies on previously published data (lifespan). Because many of these claims derive from previously published data, and the current manuscript is an extension of previously published work. The authors need to be more specific as to the actual data they present (they only use the B16 melanoma model) and the actual novelty of this manuscript.

Especially experiments on life span are published and not sufficiently addressed in this actual paper, as the title would suggest.

Reviewer #2 (Public Review):

The manuscript by Wang et al. follows up on the group's previous publication on KLF1 (EKLF) K47R mice and reduced susceptibility to tumorigenesis and increased life span (Shyu et al., Adv Sci (Weinh). Sep 2022;9(25):e2201409. doi:10.1002/advs.202201409). In the current manuscript, the authors have described the dependence of these phenotypes on age, gender, genetic background, and hematopoietic translation of bone marrow mononuclear cells. Considering the current study is centered on the phenotypes described in the previous study, the novelty is diminished. Further, there are significant conceptual concerns in the study that make the inferences in the manuscript far less convincing. Major concerns are listed below:

1. The authors mention more than once in the manuscript that KLF1 is expressed in range of blood cells including hematopoietic stem cells, megakaryocytes, T cells and NK cells. In the case of megakaryocytes, studies from multiple labs have shown that while EKLF is expressed megakaryocyte-erythroid progenitors, EKLF is important for the bipotential lineage decision of these progenitors, and its high expression promotes erythropoiesis, while its expression is antagonized during megakaryopoiesis. In the case of HSCs, the authors reference to their previous publication for KLF1's expression in these cells- however, in this study nor in the current study, there is no western blot documented to convincingly show that KLF1 protein is expressed at detectable levels in these cells. For T cells, the authors have referenced a study which is based on ectopic expression of KLF1. For NK cells, the authors reference bioGPS: however, upon inspection, this is also questionable.

2. The current study rests on the premise that KLF1 is expressed in HSCs, NK cells and leukocytes, and the references cited are not sufficient to make this assumption, for the reasons mentioned in the first point. Therefore, the authors will have to show both KLF1 mRNA and protein levels in these cells, and also compare them to the expression levels seen in KLF1 wild type erythroid cells along with knockout erythroid cells as controls, for context and specificity.

3. To get to the mechanism driving the reduced susceptibility to tumorigenesis and increased life span phenotypes in EKLF K74R mice, the authors report some observations- However, how these observations are connected to the phenotypes is unclear.
a. For example, in Figure S3, they report that the frequency of NK1.1+ cells is higher in the mutant mice. The significance of this in relation to EKLF expression in these cells and the tumorigenesis and life span related phenotypes are not described. Again, as mentioned in the second point, KLF1 protein levels are not shown in these cells.
b. In Figure 4, the authors show mRNA levels of immune check point genes, PD-1 and PD-l1 are lower in EKLF K74R mice in PB, CD3+ T cells and B220+ B cells. Again, the questions remain on how these genes are regulated by EKLF, and whether and at what levels EKLF protein is expressed in T cells and B cells relative to erythroid cells. Further, while the study they reference for EKLF's role in T cells is based on ectopic expression of EKLF in CD4+ T cells, in the current study, CD3+ T cells are used. Also, there are no references for the status of EKLF in B cells. These details are not discussed in the manuscript.

4. The authors perform comparative proteomics in the leukocytes of EKLF K74R and WT mice as shown in Figure S5. What is the status of EKLF levels in the mutant lysate vs wild type lysates based on this analysis? More clarity needs to be provided on what cells were used for this analysis and how they were isolated since leukocytes is a very broad term.

5. In the discussion the authors make broad inferences that go beyond the data shown in the manuscript. They mention that the tumorigenesis resistance and long lifespan is most likely due to changes in transcription regulatory properties and changes in global gene expression profile of the mutant protein relative to WT leukocytes. And based on reduced mRNA levels of Pd-1 Pd-l1 genes in the CD3+ T cells and B220+ B cells from mutant mice, they "assert" that EKLF is an upstream regulator of these genes and regulates the transcriptomes of a diverse range of hematopoietic cells. The lack of a ChIP assay to show binding of WT EKLF on genes in these cells and whether this binding is reduced or abolished in the mutant cells, make the above statements unsubstantiated.

6. Where westerns are shown, the authors need to show the molecular weight ladder, and where qPCR data are shown for EKLF, it will be helpful to show the absolute levels and compare these levels to those in erythroid cells, along the corresponding EKLF knock out cells as controls.

7. Figure S1D does not have a figure legend. Therefore, it is unclear what the blot in this figure is showing. In the text of the manuscript where they reference this figure, they mention that the levels of the mutant EKLF vs WT EKLF does not change in peripheral blood, while in the figure they have labeled WBCs for the blot, and the mRNA levels shown do seem to decrease in the mutant compared to WT peripheral blood.

Reviewer #3 (Public Review):

Hung et al provide a well-written manuscript focused on understanding how Eklf mutation confers anticancer and longevity advantages in vivo. The work is fundamental and the data is convincing although several details remain incompletely elucidated. The major strengths of the manuscript include the clarity of the effect and the appropriate controls. For instance, the authors query whether Eklf (K74R) imparts these advantages in a background, age, and gender dependent manner, demonstrating that the findings are independent. In addition, the authors demonstrate that the effect is not the consequence of the specific amino acid substitution, with a similar effect on anticancer activity. Furthermore, the authors provide some evidence that PD-1 and PDL-1 are altered in Eklf (K74R) mice.

Finally, they demonstrate that the effects are transferrable with BMT. Several weaknesses are also evidence. For instance, only melanoma is tested as a model of cancer such that a broad claim of "anti-cancer activity" may be somewhat of an overreach. It is also unclear why a homozygous mutation is needed when only a small fraction of cells during BMT can confer benefit. It is also difficult to explain how transplanted donor Eklf (K74R) HSCs confer anti-melanoma effect 7 and 14 days after BMT. Furthermore, it would be useful to see whether there are virulence marker alterations in the melanoma loci in WT vs Eklf (K74R) mice. Finally, the data in Fig 4c is difficult to interpret as decreased PD-1 and PDL-1 after knockdown of EKLF in vitro is not a useful experiment to corroborate how mutation without changing EKLF expression impacts immune cells. The work is impactful as it provides evidence that healthspan and lifespan may be modulated by specific hematological mutation but the mechanism by which this occurs is not completely elucidated by this work.

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