Abstract
How hematopoietic stem cells (HSCs) maintain metabolic homeostasis to support tissue repair and regeneration throughout the lifespan is elusive. Here, we show that CD38, an NAD+-dependent metabolic enzyme, promotes HSC proliferation by inducing mitochondrial Ca2+ influx and mitochondrial metabolism in young mice. Conversely, aberrant CD38 upregulation during aging is a driver of HSC deterioration in aged mice due to dysregulated NAD+ metabolism and compromised mitochondrial stress management. The mitochondrial calcium uniporter, a mediator of mitochondrial Ca2+ influx, also supports HSC proliferation in young mice yet drives HSC decline in aged mice. Pharmacological inactivation of CD38 reverses HSC aging and the pathophysiological changes of the aging hematopoietic system in aged mice. Together, our study highlights an NAD+ metabolic checkpoint that balances mitochondrial activation to support HSC proliferation and mitochondrial stress management to enhance HSC self-renewal throughout the lifespan, and links aberrant Ca2+ signaling to HSC aging.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
Sequencing data are deposited to the Gene Expression Omnibus under accession no. GSE268310. All data supporting the findings are available from the corresponding author. Source data are provided with this paper.
References
Linton, P. J. & Dorshkind, K. Age-related changes in lymphocyte development and function. Nat. Immunol. 5, 133–139 (2004).
Mohrin, M. et al. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science 347, 1374–1377 (2015).
Nakamura-Ishizu, A., Ito, K. & Suda, T. Hematopoietic stem cell metabolism during development and aging. Dev. Cell 54, 239–255 (2020).
Rossi, D. J., Jamieson, C. H. M. & Weissman, I. L. Stems cells and the pathways to aging and cancer. Cell 132, 681–696 (2008).
Mohrin, M. & Chen, D. The mitochondrial metabolic checkpoint and aging of hematopoietic stem cells. Curr. Opin. Hematol. 23, 318–324 (2016).
Wang, Y., Barthez, M. & Chen, D. Mitochondrial regulation in stem cells. Trends Cell Biol. https://doi.org/10.1016/j.tcb.2023.10.003 (2023).
Brown, K. et al. SIRT3 reverses aging-associated degeneration. Cell Rep. 3, 319–327 (2013).
Girotra, M. et al. Induction of mitochondrial recycling reverts age-associated decline of the hematopoietic and immune systems. Nat. Aging 3, 1057–1066 (2023).
Ito, K. et al. Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science 354, 1156–1160 (2016).
Luo, H. et al. Mitochondrial stress-initiated aberrant activation of the NLRP3 inflammasome regulates the functional deterioration of hematopoietic stem cell aging. Cell Rep. 26, 945–954 (2019).
Vannini, N. et al. The NAD-booster nicotinamide riboside potently stimulates hematopoiesis through increased mitochondrial clearance. Cell Stem Cell 24, 405–418 (2019).
He, M. et al. An acetylation switch of the NLRP3 inflammasome regulates aging-associated chronic inflammation and insulin resistance. Cell Metab. 31, 580–591 (2020).
Shin, J. et al. SIRT7 represses Myc activity to suppress ER stress and prevent fatty liver disease. Cell Rep. 5, 654–665 (2013).
Wang, C.-L. et al. The mitochondrial unfolded protein response regulates hippocampal neural stem cell aging. Cell Metab. 35, 996–1008 (2023).
Horenstein, A. L. et al. The circular life of human CD38: from basic science to clinics and back. Molecules 25, 4844 (2020).
Howard, M. et al. Formation and hydrolysis of cyclic ADP-ribose catalyzed by lymphocyte antigen CD38. Science 262, 1056–1059 (1993).
Chini, E. N. & De Toledo, F. G. S. Nicotinic acid adenine dinucleotide phosphate: a new intracellular second messenger? Am. J. Physiol. Cell Physiol. 282, C1191–C1198 (2002).
Deaglio, S., Vaisitti, T., Aydin, S., Ferrero, E. & Malavasi, F. In-tandem insight from basic science combined with clinical research: CD38 as both marker and key component of the pathogenetic network underlying chronic lymphocytic leukemia. Blood 108, 1135–1144 (2006).
Jin, D. et al. CD38 is critical for social behaviour by regulating oxytocin secretion. Nature 446, 41–45 (2007).
Leleu, X. et al. Anti-CD38 antibody therapy for patients with relapsed/refractory multiple myeloma: differential mechanisms of action and recent clinical trial outcomes. Ann. Hematol. 101, 2123–2137 (2022).
Tarragó, M. G. et al. A potent and specific CD38 inhibitor ameliorates age-related metabolic dysfunction by reversing tissue NAD+ decline. Cell Metab. 27, 1081–1095 e1010 (2018).
Novelli, E. M., Ramirez, M. & Civin, C. I. Biology of CD34+CD38− cells in lymphohematopoiesis. Leuk. Lymphoma 31, 285–293 (1998).
Zhao, J. L. et al. Conversion of danger signals into cytokine signals by hematopoietic stem and progenitor cells for regulation of stress-induced hematopoiesis. Cell Stem Cell 14, 445–459 (2014).
Wilkinson, A. C., Ishida, R., Nakauchi, H. & Yamazaki, S. Long-term ex vivo expansion of mouse hematopoietic stem cells. Nat. Protoc. 15, 628–648 (2020).
Mohrin, M., Widjaja, A., Liu, Y., Luo, H. & Chen, D. The mitochondrial unfolded protein response is activated upon hematopoietic stem cell exit from quiescence. Aging Cell 17, e12756 (2018).
Randall, T. D., Lund, F. E., Howard, M. C. & Weissman, I. L. Expression of murine CD38 defines a population of long-term reconstituting hematopoietic stem cells. Blood 87, 4057–4067 (1996).
Suda, T., Takubo, K. & Semenza, G. L. Metabolic regulation of hematopoietic stem cells in the hypoxic niche. Cell Stem Cell 9, 298–310 (2011).
Umemoto, T., Hashimoto, M., Matsumura, T., Nakamura-Ishizu, A. & Suda, T. Ca2+-mitochondria axis drives cell division in hematopoietic stem cells. J. Exp. Med. 215, 2097–2113 (2018).
Kirichok, Y., Krapivinsky, G. & Clapham, D. E. The mitochondrial calcium uniporter is a highly selective ion channel. Nature 427, 360–364 (2004).
Sun, X. et al. Nicotinamide riboside attenuates age-associated metabolic and functional changes in hematopoietic stem cells. Nat. Commun. 12, 2665 (2021).
Covarrubias, A. J., Perrone, R., Grozio, A. & Verdin, E. NAD+ metabolism and its roles in cellular processes during ageing. Nat. Rev. Mol. Cell Biol. 22, 119–141 (2021).
He, H. et al. Aging-induced MCPH1 translocation activates necroptosis and impairs hematopoietic stem cell function. Nat. Aging 4, 510–526 (2024).
He, H. et al. Age-related noncanonical TRMT6-TRMT61A signaling impairs hematopoietic stem cells. Nat. Aging 4, 213–230 (2024).
Hong, T. et al. TET2 modulates spatial relocalization of heterochromatin in aged hematopoietic stem and progenitor cells. Nat. Aging 3, 1387–1400 (2023).
Konturek-Ciesla, A., Olofzon, R., Kharazi, S. & Bryder, D. Implications of stress-induced gene expression for hematopoietic stem cell aging studies. Nat. Aging 4, 177–184 (2024).
Wendorff, A. A. et al. Epigenetic reversal of hematopoietic stem cell aging in Phf6-knockout mice. Nat. Aging 2, 1008–1023 (2022).
Sun, D. et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 14, 673–688 (2014).
Ohkubo, R. et al. The hepatic integrated stress response suppresses the somatotroph axis to control liver damage in nonalcoholic fatty liver disease. Cell Rep. 41, 111803 (2022).
Ibneeva, L. et al. CD38 promotes hematopoietic stem cell dormancy. PLoS Biol. 22, e3002517 (2024).
Camacho-Pereira, J. et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab. 23, 1127–1139 (2016).
Acknowledgements
This study was supported by National Institutes of Health grant nos. R01AG082105, R01DK 117481, R01AG063404 and R01AG 063389 to D.C., no. R01 AA029124 to C.J., nos. R01 AG26094, R01 AG58812 and R01 CA233790 to E.N.C.; the Glenn Foundation for Medical Research via the Paul F. Glenn Laboratories for the Biology of Aging at the Mayo Clinic (E.N.C.); the National Institute of Food and Agriculture to D.C.; the Dr. and Mrs. James C.Y. Soong Fellowship (W.-C.M.); the Taiwan Government for Study Abroad Scholarship (W.-C.M.); the QB3 Frontiers in Medical Research Fellowship (W.-C.M.); the ITO Scholarship (A.M.); and the JASSO Graduate Scholarship for Degree Seeking Students (A.M.).
Author information
Authors and Affiliations
Contributions
D.C. conceived the study. Z.S., Y.F., C.-L.W., Y.W., M.B., A.M., J.G. and F.Y. characterized the CD38 KO mice, CD38-overexpressing mice, CD38hi versus CD38lo HSC comparison, and young and old HSC comparison. S.H.P. and C.J. performed the metabolomics studies. W.-C.M. and Z.S. characterized the MCU KO mice. A.W.L. and K.H. assisted with cell sorting. C.C.S.C. and E.N.C. provided the CD38 KO mice and advised on CD38. D.C. wrote the manuscript with contributions from all authors.
Corresponding author
Ethics declarations
Competing interests
E.N.C. holds a patent on the use of CD38 inhibitors for metabolic diseases that is licensed by Elysium Health. E.N.C. is a consultant for TeneoBio, Calico, Mitobridge and Cytokinetics. E.N.C. is on the advisory board of Eolo Pharma. E.N.C. owns stocks in TeneoBio. Research in the Chini laboratory has been conducted in compliance with the Mayo Clinic conflict of interest policies. The other authors declare no competing interests.
Peer review
Peer review information
Nature Aging thanks Nicola Vannini and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 CD38 is not required for HSC maintenance under homeostatic condition at young age.
a-i. Comparison of 3-month-old WT and CD38 KO mice. A. Gating strategy for HSCs. Enriched HSCs: Lin−c-Kit+Sca1+. Highly enriched HSCs: Lin−c-Kit+Sca1+CD150+CD48−. B, C. The number of enriched (p = 0.4210) (B) and highly enriched (p = 0.4955) (C) HSCs in the bone marrow were analyzed via flow cytometry. n = 6. D, E. Ki-67 (p = 0.2928) (D) and 7-AAD (p = 0.2349) (E) staining of HSCs were analyzed via flow cytometry. n = 6. F. Lineage differentiation in the peripheral blood was analyzed via flow cytometry. MNCs, mononuclear cells (p = 0.4013). n = 5 and 6. G, H. Complete blood count analyses (G, p = 0.4174; H, p = 0.1768). n = 6. I. Bone marrow cellularity normalized by body weight (p = 0.0718). n = 6 and 5. Data are presented as mean values +/- SE. ns: p > 0.05. One-sided student’s t test.
Extended Data Fig. 2 CD38 regulates Ca2+ signaling and mitochondrial activity upon HSC proliferation.
a. HSCs isolated from 3-month-old WT and CD38 KO mice were cultured in the presence of PVA for 7 days. Total cell number was counted by hemocytometer (p = 0.0026). n = 9. b. Flow cytometry analyses of Ki-67 staining of HSCs isolated from 3-month-old WT and CD38 KO mice 16 hours post pIpC treatment (p = 0.0229). n = 6. c. HSCs isolated from 3-month-old CD38 KO mice were transduced with lentivirus overexpressing CD38 or control lentivirus and were used as donors in competitive transplantation. The percentage of total donor-derived cells in the peripheral blood of the recipients was analyzed by flow cytometry (4 weeks p = 0.0652; 8 weeks p = 0.0059; 12 weeks p = 0.0085; 16 weeks p = 0.0260). n = 12. d-n. Noncompetitive transplantation was performed using bone marrow cells from 3-month-old WT and CD38 KO mice as donors. Donor-derived enriched HSCs from the recipient mice were compared. d, e. Relative abundances of metabolites were measured by LC-MS (D, p = 0.0273; E, p = 0.0105). n = 6 and 7(D). n = 10 (E). f. ATP levels (p = 0.0007). n = 4. g-n. Representative histograms and quantification for Fluo-4 (p = 0.0357) (G, H), Rhod-2 (p = 0.0291) (I, J), TMRM (p = 0.0389) (K, L), and ROS (p = 0.0367) (M, N) staining analyzed via flow cytometry. n = 6 and 5 (G, H). n = 5 and 6 (I, J), n = 6 (K, L), n = 5 (M, N). Data are presented as mean values +/- SE. *: p < 0.05. **: p < 0.01. ***: p < 0.001. One-sided student’s t test.
Extended Data Fig. 3 CD38 regulates Ca2+ signaling and mitochondrial activity upon HSC proliferation.
a–c. Flow cytometry analyses of Rhod-2 (p = 0.0049) (A), TMRM (p = 0.0400) (B), ROS (p = 0.0497) (C) staining of HSCs isolated from 3-month-old WT and CD38 KO mice 16 hours post pIpC treatment. n = 5 and 6 (A, C). n = 5 (B). d. Gating strategy. e, f. Flow cytometry analyses of TMRM (p = 0.0265) (E) and ROS (p = 0.0137) (F) staining of CD38high and CD38low HSCs isolated from 3-month-old WT mice cultured for 18 hours in the presence of cytokines. n = 6 and 5 (E). n = 6 (F). Data are presented as mean values +/- SE. *: p < 0.05. **: p < 0.01. One-sided student’s t test.
Extended Data Fig. 4 Effect of CD38 on Ca2+ signaling and mitochondrial activity under homeostatic condition.
a-h. Comparison of enriched HSCs derived from 3-month-old WT and CD38 KO mice. A, B. Representative histograms (A) and quantification (p = 0.3041) (B) for Fluo-4 staining analyzed via flow cytometry. MFI, mean fluorescence intensity. n = 6. c, d. Representative histograms (C) and quantification (D) for Rhod-2 staining analyzed via flow cytometry (p = 0.2955). n = 6. e, f. Representative histograms (E) and quantification (F) for ROS staining analyzed via flow cytometry (p = 0.3350). n = 6. g, h. Representative histograms (G) and quantification (H) for TMRM staining analyzed via flow cytometry (p = 0.1819). n = 6. Data are presented as mean values +/- SE. ns: p > 0.05. One-sided student’s t test.
Extended Data Fig. 5 MCU is not required for HSC maintenance under homeostatic condition at young age.
a-d. Comparison of 7-month-old WT and MCU KO mice. A. HSC number in the bone marrow analyzed via flow cytometry (p = 0.2488). n = 6. B. 7-AAD staining of HSCs analyzed via flow cytometry (p = 0.2157). n = 6. C. Lineage differentiation in the peripheral blood analyzed via flow cytometry. MNCs, mononuclear cells (p = 0.2453). n = 10 and 11. D. Bone marrow cellularity normalized by body weight (p = 0.1160). n = 6. Data are presented as mean values +/- SE. ns: p > 0.05. One-sided student’s t test.
Extended Data Fig. 6 MCU is required to support HSC proliferation.
a, b. Competitive transplantation using HSCs isolated from 7-month-old WT and MCU KO mice as donors. The percentage of donor-derived HSCs in the bone marrow of the recipients (p = 0.0267) (A) and the percentage of donor-derived cells in the peripheral blood of the recipients (4 weeks p = 0.0425; 8 weeks p = 0.0101; 12 weeks p = 0.0142; 16 weeks p = 0.0090) (B) were quantified by flow cytometry. n = 6 (A). n = 14 (B). c-g. Enriched HSCs isolated from 3-month-old WT and MCU KO mice were cultured in the presence of cytokines. BrdU incorporation (p = 0.0477) (C), 7-AAD (p = 0.0984) (D), Rhod-2 (p = 0.0031) (E), TMRM (p = 0.0445) (F), and ROS (p = 0.0278) (G) staining of HSCs were analyzed by flow cytometry. n = 6 (C, D). n = 5 (E-G). Data are presented as mean values +/- SE. *: p < 0.05. **: p < 0.01. ns: p > 0.05. One-sided student’s t test.
Extended Data Fig. 7 CD38 regulates HSC function via Ca2+ signaling and mitochondrial activity.
a-d. HSCs from 3-month-old WT and CD38 KO mice were treated with or without MCU shRNA for 48 hours in culture in the presence of cytokines before flow cytometry analyses of Rhod2 staining (Ctrl, p = 0.0239; MCU shRNA, p = 0.4907) (A), TMRM staining (Ctrl, p = 0.0334; MCU shRNA, p = 0.0720) (B), HSC number (Ctrl, p = 0.0002; MCU shRNA, p = 0.2252) (C), and colony forming assay (Ctrl, p = 0.0250; MCU shRNA, p = 0.2180) (D). n = 3. Data are presented as mean values +/- SE. *: p < 0.05. ***: p < 0.001. ns: p > 0.05. One-sided student’s t test.
Extended Data Fig. 8 CD38 is upregulated in HSCs during aging.
a. Schematic illustration of the NAD+ metabolic pathways. b-g. Comparison of HSCs isolated from young (3 months old) and old (24 months old) WT mice. B. Quantitative real-time PCR analyses of NAD+ metabolic enzymes (PARP1, p = 0.4431; NAMPT, p = 0.1517; NAPRT, p = 0.1546; NMNAT3, p = 0.3542; QPRT, p = 0.0419; CD38, p = 0.0010). n = 6 and 7 (CD38). n = 3 and 4 (other enzymes). C, D. Representative histograms (C) and quantification (D) for CD38 staining analyzed via flow cytometry (p = 0.0472). n = 4. E-G. Flow cytometry analyses of Fluo-4 (p = 0.0269) (E), TMRM (p = 0.0005) (F), and ROS (p = 0.0001) (G) staining. n = 5. Data are presented as mean values +/- SE. *: p < 0.05. **: p < 0.01. ***: p < 0.001. ns: p > 0.05. One-sided student’s t test.
Extended Data Fig. 9 CD38 inactivation prevents HSC aging.
a-h. Comparison of HSCs isolated from 24-month-old WT and CD38 KO mice. A, B. RNA-sequencing analyses of enriched HSCs. Volcano plot summarizing the differentially expressed genes (Red: upregulated in CD38 KO. Blue: downregulated in CD38 KO. P adj<0.1. Benjamini-Hochberg procedure). Genes related to ribosomal proteins are labeled (A). Gene ontology analysis for the biological functions of differentially expressed genes (B). n = 2. C, D. Flow cytometry analyses of TMRM (p = 0.0076) (C) and ROS (p = 0.0058) (D) staining. n = 10 (C). n = 10 and 12 (D). E. Quantitative real-time PCR analysis of HSP60 (p = 0.0022). n = 3. F-H. Staining for 7-AAD (p = 0.0029) (F), activated caspase 1 (p = 0.0254) (G), and Ki-67 (p = 0.1974) (H) were analyzed by flow cytometry. n = 6 (F). n = 6 and 5 (G). n = 5 and 6 (H). Data are presented as mean values +/- SE. *: p < 0.05. **: p < 0.01. ns: p > 0.05. One-sided student’s t test.
Extended Data Fig. 10 MCU inactivation prevents HSC aging.
a-e. Comparison of 24-month-old WT and MCU KO mice. A, B. Complete blood count analyses (A, p = 0.0190; B, p = 0.0122). n = 8. C. Bone marrow cellularity normalized by body weight (p = 0.0181). n = 7 and 8. D, E. Competitive transplantation using HSCs isolated from 24-month-old WT and MCU KO mice as donors. The percentage of donor-derived cells in the peripheral blood of the recipients (4 weeks p = 0.0419; 8 weeks p = 0.0001; 12 weeks p = 0.0023; 16 weeks p = 0.0010) (D) and donor-derived lineage differentiation in the peripheral blood of the recipients (p = 0.0002) (E) were determined by flow cytometry. n = 12 and 9. f. A working model. CD38 catalyzes synthesis of Ca2+ second messengers from NAD+. At young age, CD38 is required for HSC proliferation through activating Ca2+ signaling and mitochondrial metabolism. NAD+ metabolism is required for activating sirtuins and suppressing mitochondrial stress. The balanced NAD+ metabolism and Ca2+ signaling ensure sufficient mitochondrial activation and adequate mitochondrial stress resistance. At old age, CD38 is upregulated, tipping the balance toward increased Ca2+ signaling and decreased NAD+ metabolism, and leading to uncontrolled mitochondrial activation and stress. Data are presented as mean values +/- SE. *: p < 0.05. **: p < 0.01. ***: p < 0.001. One-sided student’s t test.
Supplementary information
Supplementary Tables 1–3
Differentially expressed genes for RNA sequencing analyses of enriched HSCs isolated from 3-month-old WT and CD38 KO mice cultured in the presence of cytokines (Supplementary Table 1), under homeostatic conditions (Supplementary Table 2) or from 24-month-old WT and CD38 KO mice under homeostatic conditions (Supplementary Table 3). The Benjamini–Hochberg procedure.
Source data
Source Data Fig. 1
Statistical source data.
Source Data Fig. 2
Statistical source data.
Source Data Fig. 3
Statistical source data.
Source Data Fig. 4
Statistical source data.
Source Data Extended Data Fig. 1
Statistical source data.
Source Data Extended Data Fig. 2
Statistical source data.
Source Data Extended Data Fig. 3
Statistical source data.
Source Data Extended Data Fig. 4
Statistical source data.
Source Data Extended Data Fig. 5
Statistical source data.
Source Data Extended Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 7
Statistical source data.
Source Data Extended Data Fig. 8
Statistical source data.
Source Data Extended Data Fig. 9
Statistical source data.
Source Data Extended Data Fig. 10
Statistical source data.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Song, Z., Park, S.H., Mu, WC. et al. An NAD+-dependent metabolic checkpoint regulates hematopoietic stem cell activation and aging. Nat Aging (2024). https://doi.org/10.1038/s43587-024-00670-8
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s43587-024-00670-8