Skip to main content

Hematopoietic stem/progenitor cell transplantation recovers immune defects and prevents lymphomas in Atm-deficient mice

Abstract

Background

Ataxia-telangiectasia (A-T) is a rare autosomal recessive multi-system and life-shortening disease, characterized by progressive cerebellar neurodegeneration, immunodeficiency, radiation sensitivity and cancer predisposition, with high incidence of leukemia and lymphoma. A-T is caused by mutations in the gene encoding for ATM protein that has a major role in maintaining the integrity of the genome. Because there are no cures for A-T, we aimed to tackle immunodeficiency and prevent cancer onset/progression by transplantation therapy.

Methods

Enriched hematopoietic stem/progenitor cells (HSPCs), collected from bone marrow of wild-type mice, were transplanted in the caudal vein of 1 month old conditioned Atm−/− mice.

Results

Genomic analyses showed that transplanted Atm positive cells were found in lymphoid organs. B cells isolated from spleen of transplanted mice were able to undergo class switching recombination. Thymocytes were capable to correctly differentiate and consequently an increase of helper T cells and TCRβhi expressing cells was observed. Protein analysis of isolated T and B cells from transplanted mice, revealed that they expressed Atm and responded to DNA damage by initiating an Atm-dependent phosphorylation cascade. Indeed, aberrant metaphases were reduced in transplanted Atm-deficient mice. Six months after transplantation, Atm−/− mice showed signs of aging, but they maintained the rescue of T cells maturation, showed DNA damage response, and prevented thymoma.

Conclusion

We can conclude that wild-type enriched HSPCs transplantation into young Atm-deficient mice can ameliorate A-T hematopoietic phenotypes and prevent tumor of hematopoietic origin.

To the Editor,

Ataxia-Telangiectasia (A-T) is a rare recessive autosomal disorder, commonly referred as genome instability syndrome and as primary immunodeficiency (PID). A-T patients suffer of neurodegeneration, aging, sterility, radiosensitivity and cancer predisposition. The cause of the disease are mutations in the ATM (ataxia telangiectasia mutated) gene that encodes for a serine/threonine kinase of the phosphatidylinositol-3-kinase related kinase (PI3KK) family, majorly involved in cell cycle regulation and DNA repair mechanisms [1]. Malignancies of hematological origin, specifically affecting B and T cell lineages, are frequent in A-T patients, with a high incidence of 21.7% by the age of 15 years [2,3,4]. The most common forms are Acute Lymphocytic Leukemias (ALL), non-Hodgkin’s and Hodgkin’s lymphomas. In older patients are observed Chronic T-Cell Leukemias (CCL) and solid tumors, including gastric, colon, pancreas, breast, liver, esophageal and basal cell carcinomas, ovarian dysgerminoma and uterine leiomyoma [3].

Currently, there is no cure for the disease and A-T patients die before their forties while treatments are only symptomatic and supportive [5,6,7]. Bone marrow heterologous transplantation is one of the therapeutic options that could tackle immunodeficiency, as it is for other PIDs, but due to the extreme radiosensitivity of the patients and non-myeloablative conditioning side effects, its use has been restricted and the outcomes are highly unpredictable [8]. Gene therapy strategies could be then a favourable alternative to the allo-transplantation, since correcting patients’ own cells will allow autologous transplantation, avoiding most of side effects such as graft vs host disease, tissue rejection, infections, and the need of Human Leukocyte Antigen (HLA)-identical related donors [9]. In this scenario, it becomes urgent to identify the critical hematopoietic cell population to be eventually engineered with Atm and transplanted to counteract immunodeficiency and hematopoietic cancer predisposition of A-T disease.

Previous works demonstrated the feasibility of bone marrow transplantation in Atm-deficient mice and the amelioration of the phenotype [10,11,12,13] and we have already proved that global Atm reactivation can revert A-T in transgenic mouse models [14]. In this study, we expand on prior research describing the transplantation and engraftment of wild-type hematopoietic stem/progenitor cells (HSPCs) into Atm−/− mice as well as their effect on immunological markers, genomic stability and DNA damage response. Using magnetic beads, we selected Lineage negative and c-Kit positive (Linc-Kit+, LK) cells from bone marrow, that are known to be hematopoietic stem and progenitor cells [15] and so B and T cell progenitors. The final enriched LK population was around 3% of harvested cells and c-Kit+ cells both Sca-1+ and Sca-1 reached around 80% of purification (Suppl. Figure 1a). This population was maintained 24 h after culture with the significant increase of c-Kit+Sca-1+ subpopulation (Suppl. Figure 1a), already known to be capable of reconstituting hematopoiesis in lethally irradiate wild-type mice [15]. A further characterization revealed that also the proportion of CD150+ hematopoietic stem cells (HSCs) was maintained, whereas a significant increase was observed in the CD48+ hematopoietic progenitor cells (HPCs) subset (Suppl. Figure 1b).

Before transplantation, we verified the efficacy of the conditioning approach on Atm−/− mice [11,12,13], and extremely decreased T cells percentage was observed in peripheral blood for up to 7 weeks (Suppl. Figure 1c).

Then, we transplanted 3 to 5 million LK-enriched cells from wild-type male donors into conditioned Atm−/− female recipients and analyzed the LK-transplanted female mice (AtmLKT) as shown in the flow chart reported in Suppl. Figure 1d.

We found substantial improvements in CD3 positive T cells and CD4 single-positive helper T cells in transplanted mice compared to Atm−/− animals in peripheral blood 4 and 7 weeks after transplantation (Fig. 1a,b). We discovered that thymus size, which is generally hypoplastic in Atm−/− mice, gained weight 7 weeks after transplantation (Fig. 1c). Flow cytometry analysis demonstrated significant increase in CD4 single-positive helper T cells and TCRβhi thymocytes in transplanted animals, both of which reached levels comparable to wild-type mice (Fig. 1d, e). Furthermore, if Atm−/− animals normally had reduced numbers of IgG1 positive B cells owing to class switch defects, B cells from 7 weeks transplanted mice showed an increase in IgG1 positive cells, equivalent to wild-type mice (Fig. 1f). We finally observed that male transplanted LK cells indeed reached lymphoid organs in Atm−/− females as indicated by the presence of the Sry male marker and the wild-type Atm coding genes (Fig. 1g). Following DNA damage, Atm protein could phosphorylate Atm targets including the transcription factor KAP1, the cell cycle checkpoint proteins Chk1 and Chk2, and the DNA double-strand breaks marker H2AX (Fig. 1h).

Fig. 1
figure 1

Restoration of T cells in Atm/ systemic blood and thymus, and B-cells in spleen after Atm+/+ LK cells transplantation. a Histograms of total, helper and cytotoxic T cells (CD3, CD4, CD8) in peripheral blood 4 weeks after transplantation. b Histograms of total, helper and cytotoxic T cells (CD3, CD4, CD8) in peripheral blood 7 weeks after transplantation. c Thymus and spleen picture and relative weight bar charts. The ratio between organ weight (g) and body weight (g) is reported. d Representative flow cytometry panels of CD4 and CD8 T cells and relative histogram of mean values of CD4 and CD8 single positive T cells are shown. e Histogram of mean values of TCRβhi expressing cells analyzed by flow cytometry. f Representative flow cytometry dot-plot of B cells class switching cultured in LPS and IL4 for 96 h and relative histogram of mean values of IgG1 expressing cells. g Representative genomic PCR analysis of mouse tissues collected 7 weeks after LKT. The Atm 200 bp band identifies the wild-type sequence of Atm gene, whereas the Atm 400 bp band identifies the knockout sequence of Atm gene. The Sry 100 bp band identifies male cells in the female background and 18 s rRNA gene is used as a housekeeping sequence. h Western blot analysis of Atm expression and DNA damage response in isolated thymocytes and splenocytes. Neocarzinostatin (NCS) was used as DNA damage inducer. Phosphorylation of KAP1, Chk1, Chk2 (upper band) and H2AX (γH2AX) indicate DNA damage response. Tubulin was used as loading control. N = 3 mice of each group. *P ≤ 0.05 and ** P ≤ 0.01

We determined that 1 × 106 Atm+/+ LK-enriched cells were sufficient for effective T cell reconstitution in blood, Atm expression and DNA damage response in thymocytes of transplanted Atm−/− mice (Atm−/−LKTLow), 7 weeks after transplantation (Suppl. Figure 2a–c). To further elucidate the relevance of LK cells in the reconstitution of the immune system of Atm-deficient mice, Lineage negative and c-Kit depleted (Linc-Kit, LK-) cells were transplanted (Suppl. Figure 3). Notably none of the phenotypes described above were rescued by transplantation of Atm+/+ LK cells, despite they reached the lymphoid organs (Suppl. Figure 3a–h), supporting previous works on the hematopoietic function of c-Kit+Sca-1+ cells [15].

Long-term study, 6 months post-transplantation, revealed an increase of lifespans and fur whitening in transplanted Atm−/− mice (Atm−/− LKTLong; Fig. 2a, b), when compared to non-transplanted Atm−/− mice that usually die at 3–4 months before showing signs of aging in the fur. Further studies will be conducted to address if Atm−/− LKTLong mice present more aging features. Notably, improvement of the immune systems in blood and thymus with tumor prevention (Fig. 2c–e), decreased chromosome breaks of cultured B cells (Fig. 2f), reduced trans-rearrangement rates in thymocytes (Fig. 2g), and correct DNA damage response in vitro (Fig. 2h) were observed in transplanted mice compared to untreated Atm−/− animals.

Fig. 2
figure 2

Extended life span and genomic instability prevention after long-term Atm+/+ LK transplantation. a Kaplan–Meier survival curve Long-rank Mantel-Cox test for Atm+/+, Atm−/− and Atm−/−LKTLong mice. The red dotted line indicates Atm−/− mice median survival (95 days; Atm+/+ N = 31; Atm−/− N = 25; Atm−/−LKTLong N = 6 **** P ≤ 0.0001). b Picture of 7 months old mice. Atm−/− LKTLong mice present different levels of white/grey fur. c Histogram of flow cytometry analysis of blood from 7 months old Atm−/−LKTLong mice, relative to Atm+/+ littermate and 2–4 months old Atm−/− tumor-free mice. d Representative thymus and spleen pictures of 7 months old mice after LKT relative to thymi from Atm+/+ littermate and 4 months old Atm−/− tumor-free mice. e Histogram of flow cytometry analysis of isolated thymocytes from 7 months old Atm−/−LKTLong mice, relative to Atm+/+ littermate and 2–4 months old Atm−/− tumor-free mice. f Representative image of FISH assay in B cell metaphases and histogram of defective B cell metaphases. In the inset example of an aberrant chromosome. In red the PNA-bio telomere probe and in blue the DAPI DNA staining. Scale bar = 2 μm. g Pictures of PCR for gamma receptor rearrangement (γ) and gamma-beta receptors trans-rearrangement (trans) using as template genomic DNA (500 ng, 100 ng, 10 ng, 1 ng) prepared from thymocytes. The ratio among trans bands and γ is reported for 10 ng dilution PCR product. Examples of short-term (Atm−/−LKT, 7 weeks) and long-term, (Atm−/−LKTLong, 7 months) LK transplantation are reported. Histogram of fold change among trans and γ ratios in thymus of Atm−/− and Atm−/−LKTLong mice; the weakest bands among different DNA amount were considered. h Representative western blot of Atm expression and DNA damage response following NCS treatment in isolated thymocytes of long-term transplanted mice. Phosphorylation of KAP1, Chk1, Chk2 (upper band) and H2AX (γH2AX) indicate DNA damage response. Tubulin was used as loading control. N = 3 mice of each group. * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001

In conclusion, our work shows the potential of Atm+/+ LK cell transplantation to counteract immune deficiency and genomic instability at least till 7 months of age, trough restoration of Non-Homologous End Joining and Homologous recombination repair, respectively, in a mouse model of the A-T disease. Moreover, this study offers the opportunity to investigate gene therapy applications in induced Pluripotent Stem (iPS) cells or HSPCs of A-T patients followed by autologous transplantation.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

A-T:

Ataxia-Telangiectasia

PID:

Primary immunodeficiency

ATM:

Ataxia telangiectasia mutated

PI3KK:

Phosphatidylinositol-3-kinase related kinase

CSR:

Class switch recombination

DSBs:

Double-strand breaks

ALL:

Acute lymphocytic leukemias

CCL:

Chronic T-Cell Leukemias

HLA:

Human leukocyte antigen

LK:

Linc-Kit+

LK-:

Linc-Kit

PNA:

Peptide nucleic acid

NCS:

Neocarzinostatin

HSPC:

Hematopoietic progenitor cell

HSC:

Hematopoietic stem cell

HPC:

Hematopoietic progenitor cells

LKT:

LK transplantation

References

  1. Shiloh Y. ATM and related protein kinases: Safeguarding genome integrity. Nat Rev Cancer. 2003;3(3):155–68.

    Article  CAS  PubMed  Google Scholar 

  2. Reina-San-Martin B, Chen HT, Nussenzweig A, Nussenzweig MC. ATM is required for efficient recombination between immunoglobulin switch regions. J Exp Med. 2004;200(9):1103–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bakhtiar S, Salzmann-Manrique E, Donath H, Woelke S, Duecker RP, Fritzemeyer S, et al. The incidence and type of cancer in patients with ataxia-telangiectasia via a retrospective single-centre study. Br J Haematol. 2021;194(5):879–87.

    Article  CAS  PubMed  Google Scholar 

  4. Bowen S, Wangsa D, Ried T, Livak F, Hodes RJ. Concurrent V(D)J recombination and DNA end instability increase interchromosomal trans-rearrangements in ATM-deficient thymocytes. Nucleic Acids Res. 2013;41(8):4535–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Rothblum-Oviatt C, Wright J, Lefton-Greif MA, McGrath-Morrow SA, Crawford TO, Lederman HM. Ataxia telangiectasia: a review. Orphanet J Rare Dis. 2016;11(1):1–21.

    Article  Google Scholar 

  6. Lavin MF, Gueven N, Bottle S, Gatti RA. Current and potential therapeutic strategies for the treatment of ataxia-telangiectasia. Br Med Bull. 2007;81–82(1):129–47.

    Article  PubMed  Google Scholar 

  7. van Os NJH, Haaxma CA, van der Flier M, Merkus PJFM, van Deuren M, de Groot IJM, et al. Ataxia-telangiectasia: recommendations for multidisciplinary treatment. Dev Med Child Neurol. 2017;59(7):680–9.

    Article  PubMed  Google Scholar 

  8. de Oliveira BSP, Putti S, Naro F, Pellegrini M. Bone marrow transplantation as therapy for ataxia-telangiectasia: a systematic review. Cancers. 2020;12(11):1–12.

    Google Scholar 

  9. Pai SY, Notarangelo LD. Congenital disorders of lymphocyte function. 7th ed. Amsterdam: Hematology Basic Principles and Practice. Elsevier Inc; 2018. p. 710–23.

    Google Scholar 

  10. Barlow C, Hirotsune S, Paylor R, Liyanage M, Eckhaus M, Collins F, et al. Atm-deficient mice: a paradigm of ataxia telangiectasia. Cell. 1996;86(1):159–71.

    Article  CAS  PubMed  Google Scholar 

  11. Bagley J, Cortes ML, Breakefield XO, Iacomini J. Bone marrow transplantation restores immune system function and prevents lymphoma in Atm-deficient mice. Blood. 2004;104(2):572–8.

    Article  CAS  PubMed  Google Scholar 

  12. Pietzner J, Baer PC, Duecker RP, Merscher MB, Satzger-prodinger C, Bechmann I, et al. Bone marrow transplantation improves the outcome of Atm-deficient mice through the migration of Atm-competent cells. Hum Mol Genet. 2013;22(3):493–507.

    Article  CAS  PubMed  Google Scholar 

  13. Duecker RP, Gronau L, Baer PC, Zielen S, Schubert R. Stem cell transplantation in ATM- deficient Mice. Front Immunol. 2021;29(12): 693897.

    Article  Google Scholar 

  14. Di Siena S, Campolo F, Gimmelli R, Di Pietro C, Marazziti D, Dolci S, Lenzi A, Nussenzweig A, Pellegrini M. Atm reactivation reverses ataxia telangiectasia phenotypes in vivo. Cell Death Dis. 2018;9(3):314.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Okada S, Nakauchi H, Nagayoshi K, Nishikawa S, Miura Y, Suda T. In vivo and in vitro stem cell function of c-kit- and Sca-1-positive murine hematopoietic cells. Blood. 1992;80(12):3044–50.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are particularly thankful to Prof Daniela Barilà Tor Vergata University, Rome, Italy and to Dr. Margherita Doria, Academic Department of Pediatrics, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy for reagents sharing and helpful suggestions, to Dr. Gerald Pfister, Head of Flow Cytometry Facility, EMBL Rome, Italy for FACS analysis support and to Prof Anna Maria Teti University of Aquila, Aquila, Italy for critical reading.

Funding

This work was supported by AIRC 2019 [IG 23329], ANAT and FESR Lazio Innova POR 2014–2020- A0375-2020–36524 to MP.

Author information

Authors and Affiliations

Authors

Contributions

B.S. and A.G. performed research, analyzed data and wrote the manuscript; S.P. and M.M. performed experiments and analyzed data; T.O. performed experiments and GD.TV. analyzed data and revised the manuscript, C.L. and F.N. contributed to the conception of the study and critically revised the manuscript, M.P. designed the research, interpreted the data and wrote the manuscript.

Corresponding author

Correspondence to Manuela Pellegrini.

Ethics declarations

Ethics approval and consent to participate

All our experimental procedures were complied with the Directive 2010/63/EU of the European Parliament on the protection of animals used for scientific purposes and were conducted with the approval of IBBC-CNR Animal Use for Research Ethic Committee and by the Italian Ministry of Health under Protocol Number 1104/2020PR.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

40164_2024_544_MOESM1_ESM.eps

Supplementary material 1. Figure 1. Enrichment of Atm+/+ LK progenitor cell population and Flow Cytometry analysis of peripheral blood in non-myeloablative conditioned Atm-/- mice. a) Flow cytometry analysis of sequential enrichment of Lin-c-Kit+ population isolated from total bone marrow of posterior legs of Atm+/+ mice. The total bone marrow cells (left panels), the cell population recovered after lineage negative selection (middle panels), and the cell population obtained after CD117 beads, either freshly isolated (T0) or cultured for 24 h (T24), (right panels) are shown. The analysis was done with anti APC-Lineage cocktail (upper panels) and with anti PE-CD117 for c-Kit and PE-Cy7 for Sca-1 (lower panels) on gated live cells. Histogram N= 5 experiments; * P≤ 0.05, ** P≤ 0.01. b) Flow cytometry analysis of HSPC population obtained after CD117 beads stained for CD150 stem cell marker and CD48 multipotent progenitor cell marker on freshly isolated (T0) or cultured for 24 h (T24) cells. Histogram N= 3experiments; * P≤ 0.05. c) Representative dot-plot of CD4 and CD8 analysis of Atm-/- before conditioning and 4, 7 weeks after conditioning. c) Column charts of helper and cytotoxic T cells (CD4, CD8) in peripheral blood 7 weeks after conditioning. N= 3 mice of each group; ** P≤ 0.01, *** P≤ 0.001. d) Schematic representation of LK transplantation and analysis.

40164_2024_544_MOESM2_ESM.eps

Supplementary material 2. Figure 2. Partial restoration of T cells in Atm-/- mice after low Atm+/+ LK cells transplantation (Atm−/−LKTLow). a) Representative genomic PCR analysis of mouse tissues collected 7 weeks after transplantation of 1x106 Atm+/+ LK progenitor cells. The Atm 200bp band identifies the wild-type sequence of Atm gene, whereas the Atm 400bp band identifies the knockout sequence of Atm gene. N=3 mice for each group. b) Histograms of mean values of CD3 positive and CD4 single positive T cells in blood are shown after 1x106 Atm+/+ LKT. * P≤ 0.05, ** P≤ 0.01. N=3 mice for each group. c) Western blot analysis of Atm expression and DNA damage response to NCS in isolated thymocytes after 1x106 Atm+/+ LKT. pKAP1 indicates DNA damage response. Tubulin was used as loading control.

40164_2024_544_MOESM3_ESM.eps

Supplementary material 3. Figure 3. Atm-/- mice do not rescue immune system after Atm+/+ LK negative cells transplantation. a) Flow cytometry dot plot of Atm+/+ LK- population recovered after lineage negative selection and CD117 beads (T0) and cultured for 24 h (T24) before transplantation in Atm-/-. The analysis was done with anti-CD117 (c-Kit) and anti-Sca-1 antibodies on gated live cells. b) Representative genomic PCR analysis of mouse tissues collected 7 weeks after transplantation of LK positive and LK negative cells. The Atm 200bp band identifies the wild-type sequence of Atm gene, whereas the Atm 400bp band identifies the knockout sequence of Atm gene. c) Western blot analysis of Atm expression and DNA damage response in isolated thymocytes. NCS was used as DNA damage inducer. pKAP1 reveals response to DNA damage. Tubulin was used as loading control. d) Dot plots and quantitative analysis of total and helper T cells (CD3, CD4, CD8) in peripheral blood 7 weeks after transplantation. e) Thymus and spleen picture and relative weights. Scale bar 0.5cm. f) Representative flow cytometry panel of CD4 and CD8 thymocytes and relative histogram of mean values of CD4 and CD8 single positive thymocytes. g) Histogram of mean values of TCRβhi expressing cells. h) Representative flow cytometry dot-plot of B cells class switching cultured with LPS and IL4 for 96 h and relative histogram of mean values of IgG1 expressing cells. N=3 mice of each group; * P≤ 0.05, ** P≤ 0.01, ***P≤ 0.001 and **** P≤ 0.0001.

Supplementary material 4. Supplemental Material and Methods.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Oliveira, B.S.P., Giovinazzo, A., Putti, S. et al. Hematopoietic stem/progenitor cell transplantation recovers immune defects and prevents lymphomas in Atm-deficient mice. Exp Hematol Oncol 13, 81 (2024). https://doi.org/10.1186/s40164-024-00544-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-024-00544-0

Keywords