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.
Older humans are prone to many pathophysiological conditions in the hematopoietic system, such as decreased competence of the adaptive immune system1. The maintenance of the hematopoietic system throughout adult life relies on the persistence of hematopoietic stem cells (HSCs). HSCs are capable of self-renewal to give rise to daughter stem cells and of differentiation to give rise to all the blood cell types, including myeloid and lymphoid lineages. The regenerative capacity of HSCs diminishes with age2–4. HSC aging is a major cause of the pathophysiological conditions in the aging hematopoietic system. Mitochondrial metabolism is essential to support HSC function5,6. Concomitant to mitochondrial activation is mitochondrial stress, which leads to loss of HSC maintenance7–11. It is unclear how HSCs balance mitochondrial activation to support proliferation and mitochondrial stress management to support self-renewal throughout the lifespan.
NAD+ is not only a coenzyme for metabolic reactions but also possesses signaling functions to regulate diverse cellular processes, including mitochondrial stress management mediated by members of the sirtuin family of NAD+-dependent deacetylases2,7,10,12–14. CD38, a transmembrane glycoprotein, catalyzes the formation of cyclic ADP-ribose (cADPR), ADP-ribose (ADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP) from NAD+ (refs. 15,16). cADPR, ADPR and NAADP mobilize Ca2+ from intracellular Ca2+ stores16,17. CD38 regulates immune responses in lymphocytes, oxytocin secretion and social behavior, metabolic homeostasis, and is used as a malignancy marker in leukemia and a target for treating multiple myeloma18–21. Its expression in HSCs is low22, suggesting that CD38 activity needs to be kept in check for HSC maintenance. In this study, using mice in which the CD38 gene was deleted, we showed that CD38 supports Ca2+ signaling, mitochondrial activity and proliferation of HSCs at a young age. However, aberrant upregulation of CD38 in HSCs during aging results in accumulation of mitochondrial stress, with profound pathophysiological changes reminiscent of HSC aging. Thus, CD38 governs an NAD+ metabolic checkpoint that balances mitochondrial activity to support HSC proliferation and mitochondrial stress management to support HSC self-renewal.
CD38 is dispensable for HSCs under homeostasis at a young age
We characterized the phenotypically defined enriched HSCs (Lin−c-Kit+Sca1+) and highly enriched HSCs (Lin−c-Kit+Sca1+CD150 +CD48−) in young (3-month-old) female CD38 knockout (KO) mice and their wild type (WT) controls. Under homeostatic condition, about 95% of HSCs are quiescent. There was no difference in the number of HSCs in the bone marrow (BM) (Extended Data Fig. 1a–c), and the percentage of Ki-67+ (Extended Data Fig. 1d) or 7-AAD+ HSCs (Extended Data Fig. 1e), indicating that CD38 inactivation does not affect HSC survival and proliferation under homeostatic condition. There was no difference in lineage differentiation in the peripheral blood (Extended Data Fig. 1f). Complete blood count analyses showed no difference (Extended Data Fig. 1g,h). BM cellularity was not altered (Extended Data Fig. 1i). Thus, CD38 is not required for HSC maintenance and hematopoiesis under homeostatic condition at a young age.
CD38 is required to support HSC proliferation at a young age
We tested whether CD38 is required for HSC activation on proliferation. To stimulate HSC proliferation, HSCs derived from young WT and CD38 KO mice were cultured in the presence of cytokines known to induce HSC proliferation and commitment for the physiological stimulation of blood production23. After 18 h of culture, there were fewer CD38 KO HSCs than WT HSCs (Fig. 1a). Bromodeoxyuridine (BrdU) labeling showed reduced proliferation for CD38 KO HSCs (Fig. 1b), while there was no difference in HSC survival, as indicated by 7-AAD staining (Fig. 1c). HSC culture in the presence of polyvinyl alcohol (PVA) allows expansion of functional HSCs over a long time24. After 7 days of culture in the presence of PVA, the number of cells derived from CD38 KO HSCs (Extended Data Fig. 2a) was reduced.
Fig. 1 |. CD38 is required to support HSC proliferation at young age.

a–c, Enriched HSCs isolated from 3-month-old WT and CD38 KO mice were cultured in the presence of cytokines. The number of HSCs (P = 0.0113) (a), BrdU incorporation (P = 0.0215) (b) and 7-AAD staining (P = 0.3356) (c) of HSCs were analyzed using flow cytometry. a,b, n = 6. c, n = 6 and 5. d–f, Competitive transplantation using HSCs isolated from 3-month-old WT and CD38 KO mice as donors. The percentage of donor-derived HSCs in the BM of the recipients (P = 0.0144) (d), donor-derived lineage differentiation in the peripheral blood (P = 0.2442) (e) and the percentage of donor-derived cells in the peripheral blood of the recipients (4 weeks, P < 0.0001; 8 weeks, P = 0.0017; 12 weeks, P = 0.0018; 16 weeks, P = 0.0039) (f) were analyzed using flow cytometry. d, n = 5 and 6. e,f, n = 11 and 12. g,h, Noncompetitive transplantation using whole-BM cells isolated from 3-month-old WT and CD38 KO mice as donors. Ki-67 staining (P = 0.0442) (g) and 7-AAD staining (P = 0.1802) (h) of donor-derived HSCs were analyzed using flow cytometry 1 month after transplantation. g, n = 5. h, n = 6. i, The homing ability of BM cells from 3-month-old WT and CD38 KO mice 16 h after transplantation was analyzed using flow cytometry (P = 0.2659). n = 6. Data are presented as the mean ± s.e. *P < 0.05, **P < 0.01, ***P < 0.001, NS P > 0.05. A one-sided Student’s t-test was used. MNC, mononuclear cell.
To stimulate HSC proliferation in vivo, we performed a competitive BM transplantation assay using HSCs isolated from young WT and CD38 KO mice as donors. Sorted HSCs from CD45.2 donor mice were mixed with CD45.1 B6.SJL competitor BM cells and were injected into lethally irradiated B6.SJL recipient mice. CD38 KO HSCs showed reduced engraftment in the BM of recipient mice (Fig. 1d), comparable lineage differentiation in peripheral blood (Fig. 1e) and reduced capacity to reconstitute the blood system of lethally irradiated recipient mice (Fig. 1f). Staining of donor-derived HSCs in the BM of recipient mice showed a reduced frequency of Ki-67+ CD38 KO HSCs (Fig. 1g), while the percentage of 7-AAD+ HSCs was comparable between the two genotypes (Fig. 1h), suggesting that reduced CD38 KO HSC engraftment was due to reduced proliferation but not cell death. There was no difference in homing ability (Fig. 1i). As an alternative approach to stimulate HSC proliferation in vivo, we treated mice with poly(I:C)25. The frequency of Ki-67+ HSCs was reduced in CD38 KO mice on poly(I:C) treatment (Extended Data Fig. 2b). Together, these data indicate that CD38 is required to support HSC proliferation at a young age. Consistently, although the CD38lo population is more enriched for colony-forming cells compared to the CD38hi population, the CD38hi population proliferates faster, forms larger colonies and supports long-term reconstitution26.
To determine whether CD38 regulates HSC function autonomously, we performed a competitive BM transplantation assay using CD38 KO HSCs with or without CD38 overexpression as donors. CD38 overexpression resulted in increased capacity to reconstitute the blood system of lethally irradiated recipient mice (Extended Data Fig. 2c), suggesting that CD38 regulates HSC function autonomously.
CD38 promotes mitochondrial activity on HSC proliferation
On cytokine stimulation, RNA sequencing (RNA-seq) analyses of HSCs isolated from young WT and CD38 KO mice showed differentially expressed genes related to cell cycle and mitochondria (Fig. 2a–c and Supplementary Table 1). In contrast, HSCs derived from young WT and CD38 KO mice under homeostatic condition did not show these changes (Supplementary Table 1). To explore the metabolic effects of CD38 in HSCs on proliferation, we administered [U-13C]glucose to WT and CD38 KO HSCs ex vivo in the presence of cytokine stimulation. Metabolomics revealed that the total levels of the tricarboxylic acid (TCA) cycle intermediates succinate and fumarate were reduced in CD38 KO HSCs (Fig. 2d,e). CD38 KO HSCs had reduced incorporation of [U-13C]glucose into the TCA cycle intermediates (Fig. 2f,g), which is consistent with reduced mitochondrial metabolism.
Fig. 2 |. CD38 regulates Ca2+ signaling and mitochondrial activity on HSC proliferation.

a–l, Enriched HSCs isolated from 3-month-old WT and CD38 KO mice were cultured in the presence of cytokines. a–c, RNA-seq analyses of enriched HSCs. a, Volcano plot summarizing differentially expressed genes (red, upregulated in CD38 KO; blue, downregulated in CD38 KO. Padj < 0.1. Benjamini–Hochberg-corrected). Genes related to HSC proliferation are labeled. b,c, Gene set enrichment analysis (GSEA) for mitochondria (b) and cell cycle (c). n = 2. d–g, [U-13C]glucose was administered to the HSC culture. Relative abundances of total metabolites (succinate, P = 0.0399 (d); fumarate, P = 0.0273 (e)) and 13C-labeled metabolites (malate, P = 0.0362 (f); fumarate, P = 0.0076 (g)) in enriched HSCs were measured using liquid chromatography–mass spectrometry (LC–MS). n = 4. h, ATP levels in enriched HSCs (P = 0.0193). n = 3. i–l, The staining for Fluo-4 (P = 0.0071) (i), Rhod-2 (P = 0.0245) (j), TMRM (P = 0.0027) (k) and ROS (P = 0.0272) (l) of HSCs was analyzed using flow cytometry. i,k,l, n = 5 and 6. j, n = 6. Data are presented as the mean ± s.e. *P < 0.05. **P < 0.01. A one-sided Student’s t-test was used. MFI, mean fluorescence intensity; NES, normalized enrichment score.
HSCs reside in a hypoxic niche in vivo and primarily rely on glycolysis for energy production27. To further explore the metabolic effects of CD38 in HSCs on proliferation in vivo, we performed metabolomics of HSCs isolated from mice transplanted with WT and CD38 KO HSCs 1 month after transplantation. The levels of fructose 6-phosphate and lactate were increased in HSCs derived from mice transplanted with CD38 KO HSCs (Extended Data Fig. 2d,e), which is consistent with reduced mitochondrial metabolism and compensatory glycolysis. Cellular ATP levels were reduced in CD38 KO HSCs on either cytokine-stimulated proliferation (Fig. 2h) or transplantation-stimulated proliferation (Extended Data Fig. 2f). Together, these data suggest that in HSCs, CD38 is required for mitochondrial metabolism and proliferation.
Mitochondrial Ca2+ influx supports HSC proliferation
On HSC transition from quiescence to proliferation, cytosolic and mitochondrial Ca2+ levels increased28. Staining with Rhod-2, a fluorescent indicator for mitochondrial Ca2+ levels, and Fluo-4, a fluorescent indicator that monitors cytoplasmic Ca2+ levels, showed that both cytosolic and mitochondrial Ca2+ levels were reduced in CD38 KO HSCs on cytokine-stimulated proliferation (Fig. 2i,j) and transplantation-stimulated proliferation (Extended Data Fig. 2g–j). Mitochondrial activity and stress, as indicated by staining for tetramethylrhodamine methyl ester (TMRM) that detects mitochondrial membrane potential and MitoSOX mitochondrial superoxide indicator, were decreased in CD38 KO HSCs under these proliferation conditions (Fig. 2k,l and Extended Data Fig. 2k–n). Additionally, CD38 KO HSCs also showed reduced Ca2+ levels, mitochondrial membrane potential and stress under poly(I:C)-induced proliferation (Extended Data Fig. 3a–c). CD38lo HSCs had lower mitochondrial membrane potential and stress than CD38hi HSCs on cytokine-stimulated proliferation (Extended Data Fig. 3d–f). In contrast, there was no difference in Ca2+ levels, mitochondrial membrane potential and stress between quiescent WT and CD38 KO HSCs under homeostatic condition (Extended Data Fig. 4a–h).
To determine whether mitochondrial Ca2+ influx is required to support HSC proliferation, a loxP allele for the mitochondrial calcium uniporter (MCU) located in the organelle’s inner membrane29, was crossed with Vav-iCre transgenic mice to delete MCU in the HSCs. HSCs in Vav-iCre+/−;MCUloxP/loxP mice (referred to as MCU KO mice below) and Vav-iCre+/− mice (referred to as WT mice below) were characterized. Under homeostatic condition, there was no difference in the number of HSCs in the BM (Extended Data Fig. 5a) and in the percentage of 7-AAD+ HSCs (Extended Data Fig. 5b). There was no difference in lineage differentiation in peripheral blood (Extended Data Fig. 5c). BM cellularity was unchanged (Extended Data Fig. 5d). Thus, MCU is not required for HSC maintenance and hematopoiesis under homeostatic condition. However, under transplantation-stimulated proliferation, MCU KO HSCs showed reduced engraftment in the BM of recipient mice (Extended Data Fig. 6a) and reduced capacity to reconstitute the blood system of lethally irradiated recipient mice (Extended Data Fig. 6b). BrdU labeling showed reduced proliferation of MCU KO HSCs compared to WT HSCs on cytokine-stimulated proliferation (Extended Data Fig. 6c). The frequency of 7-AAD+ HSCs was comparable between the two genotypes (Extended Data Fig. 6d). MCU KO HSCs had reduced Ca2+ signaling (Extended Data Fig. 6e), mitochondrial membrane potential (Extended Data Fig. 6f) and stress (Extended Data Fig. 6g). Together, these data suggest that mitochondrial Ca2+ influx is required for HSC proliferation.
To determine whether CD38 regulates HSC function by Ca2+ signaling and mitochondrial activation, we cultured HSCs from 3-month-old WT and CD38 KO mice treated with or without MCU short hairpin RNA (shRNA) for 2 days in the presence of cytokines. While CD38 KO HSCs showed reduced mitochondrial Ca2+ levels, mitochondrial membrane potential, number of HSCs and colony-forming ability compared to WT HSCs in the absence of MCU shRNA, these differences were blunted in the presence of MCU shRNA (Extended Data Fig. 7a–d), suggesting that CD38 regulates HSC function via Ca2+ signaling and mitochondrial activation.
CD38 is upregulated in HSCs during aging
NAD+ levels are reduced in HSCs during aging30. The cellular NAD+ pool is maintained by several biosynthesis pathways from NAD+ precursors (the Preiss–Handler, de novo biosynthesis and salvage pathways) (Extended Data Fig. 8a)31. The major NAD+-consuming enzymes include sirtuins, poly (ADP-ribose) polymerases (PARPs) and CD38. We examined the expression of NAD+ metabolic enzymes in HSCs of young (3-month-old) and aged (24-month-old) WT mice. The expression of several enzymes for NAD+ biosynthesis (NAMPT, NAPRT, NMNAT3) and consumption (PARP1) was unchanged (Extended Data Fig. 8b). The expression of several sirtuins was reduced in aged HSCs2,7,10, while the expression of quinolinate phosphoribosyltransferase (QPRT) was slightly increased in aged HSCs (Extended Data Fig. 8b). Because sirtuins are NAD+-consuming enzymes and QPRT is a key enzyme in de novo NAD+ biosynthesis, these changes are unlikely to contribute to the reduced NAD+ levels during aging. The expression of CD38, an NAD+-consuming enzyme, was increased in aged HSCs (Extended Data Fig. 8b–d), suggesting that increased CD38 expression may contribute to a decline in NAD+ levels in HSCs during aging. Aged HSCs had increased Ca2+ signaling (Extended Data Fig. 8e), reduced mitochondrial membrane potential (Extended Data Fig. 8f) and increased mitochondrial stress (Extended Data Fig. 8g) on cytokine-stimulated proliferation, which is consistent with CD38 activation during HSC aging.
CD38 upregulation is a driver of HSC aging
We sought to determine whether CD38 upregulation is a driver of HSC aging. HSC aging is characterized by myeloid-biased differentiation, reduced regenerative capacity per cell and defective hematopoiesis2,32–36. Compared to age-matched WT controls, aged (24-month-old) CD38 KO mice had increased NAD+ levels in HSCs (Fig. 3a), increased white blood cell count and lymphocyte count in the peripheral blood (Fig. 3b,c), and alleviated myeloid-biased differentiation in peripheral blood (Fig. 3d). BM cellularity was increased in aged CD38 KO mice (Fig. 3e). In a competitive transplantation assay, HSCs from aged CD38 KO mice exhibited improved engraftment in the BM of recipient mice (Fig. 3f) and improved regenerative capacity to reconstitute the blood system of lethally irradiated mice (Fig. 3g). The recipient mice of old CD38 KO HSCs showed ameliorated donor-derived, myeloid-biased differentiation in peripheral blood (Fig. 3h). Thus, aged CD38 KO mice are protected from HSC aging. RNA-seq analyses of HSCs derived from aged WT and CD38 KO mice showed reduced expression of ribosomal proteins in aged CD38 KO HSCs (Extended Data Fig. 9a,b and Supplementary Table 1). Increased expression of ribosomal proteins is a molecular signature of HSC aging37. Therefore, reduced expression of ribosomal proteins in aged CD38 KO HSCs is consistent with the reversal of HSC aging.
Fig. 3 |. CD38 inactivation prevents HSC aging.

a–h, Comparison of 24-month-old WT and CD38 KO mice. a, NAD+ levels in enriched HSCs (P = 0.0454). n = 5 and 6. b,c, White blood cell (WBC) (b) and lymphocyte (c) analyses (P = 0.0182 (b); P = 0.0193 (c)). n = 8 and 7. d, Lineage differentiation in peripheral blood was analyzed using flow cytometry (P = 0.0392). n = 6 and 7. e, BM cellularity normalized according to body weight (P = 0.0472). n = 6 and 8. f–h, Competitive transplantation using HSCs isolated from 24-month-old WT and CD38 KO mice as donors. Donor-derived HSC engraftment in the BM (P = 0.0311) (f), the percentage of total donor-derived cells in peripheral blood (4 weeks, P = 0.0647; 8 weeks, P = 0.0313; 12 weeks, P = 0.0090; 16 weeks, P = 0.0104) (g) and donor-derived lineage differentiation in the peripheral blood (P = 0.0107) (h) of the recipients were analyzed using flow cytometry. f, n = 6. g,h, n = 12 and 14. Data are presented as the mean ± s.e. *P < 0.05. **P < 0.01. A one-sided Student’s t-test was used.
We further took a pharmacological approach to determine the role of CD38 in HSC aging. We treated 24-month-old WT mice with 78c, a small-molecule inhibitor of CD38 (refs. 21,38), for 8 weeks (Fig. 4a). Treatment with 78c increased the NAD+ levels in HSCs (Fig. 4b), increased the white blood cell and lymphocyte count in peripheral blood to the levels equivalent to those of young mice (Fig. 4c–h), increased BM cellularity (Fig. 4i), reversed myeloid-biased differentiation in peripheral blood (Fig. 4j), decreased the frequency of myeloid-biased HSCs in the BM (Fig. 4k) and increased the number of colony-forming cells in the BM (Fig. 4l,m). The mitochondrial membrane potential and stress in HSCs were reduced by 78c treatment (Fig. 4n,o). Collectively, these data indicate that CD38 upregulation drives HSC aging and the pathophysiological changes of the aging hematopoietic system.
Fig. 4 |. 78c treatment reverses HSC aging.

a–o, Twenty-four-month-old WT mice were treated with or without 78c for 8 weeks. a, Schematic illustration of the experimental setup and timeline. b, NAD+ levels in enriched HSCs (P = 0.0002). n = 6. c–h, Lymphocyte and WBC analyses of young and aged mice before treatment (c,f) (P = 0.0009 (c); P = 0.0561 (f)) and aged mice after treatment for 5 weeks (d,g) (P = 0.0387 (d); P = 0.0888 (g)) or 8 weeks (e,h) (P = 0.0142 (e); P = 0.0459 (h)). c,f, n = 10 and 25. d,g, n = 9. e,h, n = 10 and 9. i, BM cellularity normalized according to body weight (P = 0.0485). n = 6. j, Lineage differentiation in peripheral blood was analyzed using flow cytometry (P = 0.0474). n = 9. k, Lineage-biased HSCs in the BM were analyzed using flow cytometry. Myeloid-biased HSCs: Lin−c-Kit+Sca1+CD135−CD34−CD150hi. Lineage-balanced HSCs: Lin−c-Kit+Sca1+CD135−CD34−CD150lo (P = 0.0029). n = 6. l,m, Colonies formed in a colony-forming assay using BM cells (l) and quantification (P = 0.0038) (m). n = 6 and 5. n,o, Staining for TMRM (P = 0.0490) (n) and ROS (P = 0.0362) (o) of HSCs was analyzed using flow cytometry. n, n = 5 and 6. o, n = 5. Data are presented as the mean ± s.e. *P < 0.05, **P < 0.01, ***P < 0.001. A one-sided Student’s t-test was used. Ctrl, control.
Alleviated mitochondrial stress in HSCs of aged CD38 KO mice
We investigated whether CD38 upregulation drives HSC aging due to reduced NAD+ metabolism. Several members of the sirtuin family of NAD +-dependent deacetylases (SIRT2, SIRT3 and SIRT7) are critical regulators of HSC aging that govern mitochondrial stress management programs and prevent HSCs from undergoing caspase 1-mediated cell death2,7,10. Aged CD38 KO HSCs showed reduced mitochondrial membrane potential and oxidative stress (Extended Data Fig. 9c,d), reduced expression of HSP60, a marker of mitochondrial protein folding stress (Extended Data Fig. 9e), reduced percentage of 7-AAD+ HSCs (Extended Data Fig. 9f), reduced percentage of activated caspase 1+ HSCs (Extended Data Fig. 9g) and comparable HSC proliferation (Extended Data Fig. 9h), which is consistent with elevated NAD+ signaling and sirtuin activities.
Protection of HSC aging in MCU KO mice
To determine the contribution of Ca2+ signaling to HSC aging, we characterized 24-month-old MCU KO mice. Aged MCU KO mice showed increased white blood cell and lymphoid cell count in peripheral blood (Extended Data Fig. 10a,b) and increased BM cellularity (Extended Data Fig. 10c). In a competitive transplantation assay, HSCs from aged MCU KO mice showed improved regenerative capacity to reconstitute the blood system of lethally irradiated recipient mice (Extended Data Fig. 10d) and ameliorated myeloid-biased differentiation (Extended Data Fig. 10e). These data suggest that reducing mitochondrial Ca2+ signaling protects against HSC aging.
Discussion
Our study established CD38 as a critical regulator of HSC fate decision balancing NAD+ metabolism and Ca2+ signaling to fine-tune mitochondrial activity and stress management (Extended Data Fig. 10f). Because female mice show a more pronounced myeloid-biased differentiation with aging, we primarily characterized female mice in this study. Mitochondrial activation and stress management were thought to be regulated independently. Our findings suggest that these two processes are more closely linked than previously thought. The existence of such a mechanism in HSCs highlights the importance of fine-tuning mitochondrial activity in HSC fate decisions. CD38 is required for Ca2+ signaling, mitochondrial activity and proliferation of HSCs at a young age. However, when CD38 is upregulated in old age, it tips the balance between Ca2+ signaling-mediated mitochondrial activity and NAD+ signaling-mediated mitochondrial stress management governed by sirtuins, leading to aging-associated HSC decline and pathophysiological changes of the aging hematopoietic system. Thus, CD38 must be kept in check to support HSC maintenance, which is in keeping with the observation that the CD38lo cell population is enriched for HSCs22.
A recent study examined CD38 in dormant HSCs39. Ibneeva et al.39 used CD38 KO mice and their results were consistent with our findings, that is, CD38 KO HSCs showed reduced reconstitution on BM transplantation and there was no difference in HSC proliferation between young WT and CD38 KO mice under homeostatic conditions. However, most experiments in the study by Ibneeva et al. were based on comparison of CD38hi and CD38lo HSCs, which may differ in more than just CD38 expression. Indeed, Ibneeva et al. showed that while there was no difference in HSC proliferation between young WT and CD38 KO mice under homeostatic conditions, CD38lo HSCs were more proliferative than CD38hi HSCs under the same condition, indicating that the difference in proliferation between CD38lo and CD38hi HSCs is not due to CD38 expression. They further showed that CD38lo HSCs were more proliferative than CD38 KO HSCs, again suggesting that increased proliferation of CD38lo HSCs is not due to CD38 expression.
The age-dependent effects of CD38 and MCU on HSCs are striking. The sharp contrast in the requirement of CD38 and MCU for supporting HSC function between young and old ages is consistent with the notion that mitochondrial metabolism is essential to support HSC proliferation; yet, increased mitochondrial stress in old HSCs drives their deterioration5,6. These observations also beg the question as to when and how the metabolic checkpoint becomes dysregulated along the lifespan. Importantly, pharmacological inhibition of CD38 reverses HSC aging and pathophysiological changes of the aging hematopoietic system, giving hope for the therapeutic potential of the NAD+ metabolic checkpoint regulating HSC activation and aging. Deletion of MCU is sufficient to prevent HSC aging, linking Ca2+ signaling to HSC aging and providing a nodal control point for intervention.
Our findings have important implications in understanding the current CD38-targeting treatments and informing potential CD38-targeting treatments. Anti-CD38 antibody therapy is transforming the treatment of multiple myeloma because of its anti-myeloma efficacy and manageable safety profile20. Our findings suggest that anti-CD38 antibody treatment could reverse HSC aging and the pathophysiological changes of the aging immune system, contributing to its anti-myeloma efficacy and safety profile. CD38 is a malignancy marker for leukemia18. Increased expression of CD38 in aged HSCs may account for increased susceptibility of older individuals to leukemia.
Methods
Mice
CD38 KO mice (C57BL/6) have been described previously40. B6.SJL-Ptprca Pepcb/BoyJ mice, Vav-iCre mice (C57BL/6) and MCUloxP/loxP mice (C57BL/6) were obtained from the Jackson Laboratory. For experiments using young and aged WT mice, C57BL/6 mice ranging from 3 to 24 months old were obtained from the National Institute on Aging. Female mice show more pronounced myeloid-biased differentiation with aging and were used in the study. All mice were housed on a 12–12 h light–dark cycle at 25 °C. Mice were provided with standard chow (PicoLab Rodent Diet 20, cat. no. 5053, LabDiet) and water ad libitum. Humidity was kept at 30–70%. On-site veterinarians provided health status checks. Animal procedures were performed in accordance with the University of California, Berkeley animal care committee. Mice were injected intraperitoneally with 5 mg kg−1 poly(I:C) (Sigma-Aldrich) and were analyzed 16 h later.
Flow cytometry and cell sorting
BM cells were obtained by crushing the long bones with staining medium (sterile PBS without calcium and magnesium supplemented with 2% FCS). Lineage staining contained a cocktail of anti-mouse antibodies against Mac-1 (CD11b), Gr-1 (Ly-6G/C), Ter119 (Ly-76), CD3, CD4, CD8a (Ly-2) and B220 (CD45R) (all BioLegend) at 4 °C for 20 min. For detection or sorting of HSCs, we used Lin-APC-Cy7, c-Kit-APC, Sca-1-Pacific Blue, CD48-FITC and CD150-PE antibodies (all BioLegend). For congenic strain discrimination, anti-CD45.1-PerCP and anti-CD45.2-PE-Cy7 antibodies (both from BioLegend) were used. Antibodies were diluted 1:100. To assess cell death and cell cycle, 7-AAD or Ki-67 (both from BioLegend) staining were performed according to the manufacturer’s recommendations after cell surface staining. For intracellular activated caspase-1 staining, fluorescently labeled inhibitors of caspase probe staining (ImmunoChemistry Technologies) was performed as described previously10.
For intracellular calcium, mitochondrial calcium, mitochondrial membrane potential and mitochondrial superoxide levels, cells were incubated with 1 μM Fluo-4, AM (Thermo Fisher Scientific), 1 μM Rhod-2, AM (Thermo Fisher Scientific), 1 μM TMRM (Thermo Fisher Scientific) or 5 μM MitoSOX Red Mitochondrial Superoxide Indicator (Thermo Fisher Scientific), respectively, for 30 min at 37 °C in the dark after HSC surface staining. Cells were washed with staining medium before flow cytometry analysis. Data were collected on a Fortessa analyzer with the FACSDiva software (BD Biosciences); data analyses were performed with FlowJo v.10.4 (FlowJo LLC). For cell sorting, c-Kit enrichment was performed according to the manufacturer’s instructions (Miltenyi Biotec). Cells were sorted using an Aria Sorter (BD Biosciences).
HSC culture
For cytokine-stimulated HSC proliferation, enriched HSCs were cultured with StemSpan SFEM (STEMCELL Technologies) supplemented with 10% ES-Cult FCS (STEMCELL Technologies), 1% penicillin-streptomycin (Invitrogen), interleukin-3 (IL-3) (20 ng ml−1), interleukin-6 (IL-6) (20 ng ml−1), thrombopoietin (TPO) (50 ng ml−1), Flt3l (50 ng ml−1) and stem cell factor (SCF) (100 ng ml−1) (PeproTech). For HSC proliferation, cells were pulsed with 10 μM BrdU (Invitrogen) for 18 h at 37 °C. BrdU-pulsed cells were resuspended in 100 μl staining medium. After HSC surface staining, cells were washed with staining medium, resuspended in 0.5 ml of fixation buffer (BioLegend) and incubated at room temperature for 20 min. Cells were washed with 1× perm/wash buffer (BioLegend). Then, 30 μl DNase I were added to 170 μl resuspended cells and incubated at 37 °C for 60 min. Cells were washed with 1× perm/wash buffer. Then, 2 μl of diluted anti-BrdU antibody (1:25 dilution of stock in 1× perm/wash buffer) was added to 100 μl of resuspended cells and incubated at room temperature for 30 min. Cells were washed with 1× perm/wash buffer before flow cytometry analysis.
Long-term HSC culture with PVA was performed as described elsewhere24. Briefly, 50 HSCs (Lin−c-Kit+Sca1+CD150+CD34−) were sorted into a fibronectin-treated 96-well plate (Corning) and cultured with F-12 medium (Thermo Fisher Scientific), 1% ITSX (Thermo Fisher Scientific), 1% P/S/G, 10 mM HEPES, 100 ng ml−1 mouse TPO, 10 ng ml−1 mouse SCF and 0.1% PVA (Sigma-Aldrich). Complete medium was changed on day 5. Cells were counted using a hemocytometer after 7 days.
Transplantation
For noncompetitive transplantation, 2 × 106 BM cells from CD45.2 donors were injected into lethally irradiated (950 Gy) CD45.1 B6.SJL recipient mice. The BM cells of recipient mice were analyzed 1 month after transplantation.
For competitive transplantation, 250 freshly sorted HSCs from CD45.2 donor mice were mixed with 5 × 105 CD45.1 B6.SJL competitor BM cells and injected into lethally irradiated B6.SJL recipient mice. To assess the multilineage reconstitution of transplanted mice, peripheral blood was collected every month for 4 months. Red blood cells were lysed using FACS lysing solution (BD Biosciences) for 5 min at room temperature, washed with PBS and stained with an antibody cocktail consisting of CD45.2-PE-Cy7, CD45.1-PerCP-Cy5.5, Mac1-PE, Gr1-FITC, B220-APC and CD3-Pacific Blue (BioLegend) for 20 min at 4 °C. Cells were washed with PBS before the flow cytometry analyses. BM cells were analyzed 4 months after transplantation.
Homing
A total of 2 × 106 BM cells from CD45.2 donors were injected into lethally irradiated (950 Gy) CD45.1 B6.SJL recipient mice; 16 h after transplantation, the BM cells of recipients were stained with CD45.2-PE-Cy7 antibody before the flow cytometry analyses.
Lentiviral production
CD38 was cloned into the pFUGw lentiviral construct. MCU shRNA was cloned into the pLKO.5 lentiviral construct (TRCN0000251263, Sigma-Aldrich). 293T cells were transfected with lentiviral packaging plasmids together with the pFUGw or pLKO.5 vectors using PEI Max (Polysciences) according to the manufacturer’s protocol. Then, 24 h after transfection, cells were changed to fresh medium. Forty-eight hours after transfection, the virus was collected and filtered through a 45-μm syringe filter. Fresh medium was added back to cells; the second round of virus collection was conducted 72 h after transfection. Lentivirus was concentrated using centrifugation at 17,900g, at 4 °C for 90 min and resuspended in HSC medium (StemSpan SFEM, STEMCELL Technologies) supplemented with 10% ES-Cult FCS (STEMCELL Technologies), 1% penicillin-streptomycin, IL-3 (20 ng ml−1), IL-6 (20 ng ml−1), TPO (50 ng ml−1), Flt3l (50 ng ml−1) and SCF (100 ng ml−1) (PeproTech).
Lentiviral transduction of HSCs
Freshly isolated HSCs were cultured with HSC medium for 3 h at 37 °C. Lentiviral medium was added to HSCs in a 96-well round-bottom plate and centrifuged for 90 min at 270g in the presence of 10 μg ml−1 polybrene. After centrifugation, the lentiviral medium was replaced by fresh HSC medium. This process was repeated 24 h later.
78c treatment
78c was administered to mice via intraperitoneal injection (10 mg kg−1 per dose) twice daily over a period of 8 weeks. Body weight was measured weekly. Control mice received vehicle (5% dimethylsulfoxide, 15% polyethylene glycol 400, 80% of 15% hydroxypropyl-γ-cyclodextrin (in citrate buffer, pH 6.0)) injection.
mRNA analysis
RNA was isolated from cells using TRIzol reagent (Invitrogen) according to the manufacturer’s instructions. Complementary DNA was generated using the qScript cDNA SuperMix (Quanta Biosciences) according to the manufacturer’s instructions. Gene expression was determined using real-time PCR with the Eva qPCR SuperMix Kit (BioChain Institute) on an ABI StepOnePlus system. The following primers were used: CD38, forward: 5′-GGACGAAACACCGGCCATGTAAC-3′; reverse: 5′-AACCTTCCCGACTTAGGGGC-3′; HSP60, forward, 5′-ACCTGTGACAACCCCTGAAG; reverse: 5′-TGACACCCTTTCTTCCAACC-3′; PARP1, forward: 5′-GGCAGCCTGATGTTGAGGT-3′; reverse: 5′-GCGTACTCCGCTAAAAAGTCAC-3′; NAMPT, forward: 5′-GCAGAAGCCGAGTTCAACATC-3′; reverse: 5′-TTTTCACGGCATTCAAAGTAGGA-3′; NAPRT, forward: 5′-TGCTCACCGACCTCTATCAGG-3′; reverse: 5′-CGAAGGAGCCTCCGAAAGG-3′; NMNAT3, forward: 5′-CCTGTGGTTCCTTCAACCCC-3′; reverse: 5′-AGATGATGCCCTCAATCACCT-3′; and QPRT, forward: 5′-CCGGGCCTCAATTTTGCATC-3′; reverse: 5′-GGTGTTAAGAGCCACCCGTT-3′.
RNA-seq analysis
The total RNA of sorted enriched HSCs was isolated with the RNeasy Plus Micro Kit (QIAGEN) according to the manufacturer’s instructions. RNA-seq libraries were prepared using QuantSeq 3′ mRNA-Seq Library Prep Kit FWD (Lexogen); sequencing was performed on an HiSeq 4000 system. Data analysis was processed within the Galaxy public server (https://usegalaxy.org/). Briefly, after removing low-quality reads and adapter sequences, data were mapped against the mouse genome (mm10) using HISAT2 (Galaxy v.2.2.1). Gene expression levels were measured from feature counts using HTSeq (Galaxy v.0.9.1). Differential gene expression was analyzed using DESeq2 (Galaxy v.1.34.0). Differentially expressed genes (Padj < 0.1, Benjamini–Hochberg-corrected) between CD38 KO and WT samples were uploaded to the online Database for Annotation, Visualization and Integrated Discovery v.6.8 (https://david.ncifcrf.gov/) for functional annotations. Differentially expressed genes were also subjected to GSEA using the GSEA v.4.3.2 software (https://www.gsea-msigdb.org/gsea/index.jsp).
Metabolomics sample preparation
In vivo.
BM cells were obtained by crushing the long bones with ice-cold sterile PBS without calcium and magnesium and were subjected to lysis using ice-cold ACK lysis buffer for 5 min on ice. This was followed by c-Kit enrichment and HSC surface staining as described above. After staining, the cell pellet was washed with ice-cold PBS and resuspended in 1 ml ice-cold saline (9 g l−1 NaCl in deionized water) to prevent phosphate-induced ion suppression in MS. The sorter was configured with a 70-μm nozzle tip and the sheath fluid consisted of 5 g l−1 NaCl in deionized water. A total of 5 × 104 enriched HSCs were sorted directly into 50 μl ice-cold acetonitrile (ACN) for rapid quenching and to extract metabolites. All procedures were carried out at 4 °C to minimize metabolic changes during sample preparation.
Ex vivo.
A total of 5 × 104 enriched HSCs were cultured in glucose-free Roswell Park Memorial Institute 1640 medium (Thermo Fisher Scientific) supplemented with 10% dialyzed FCS (Cytiva), 1% penicillin-streptomycin, IL-3 (20 ng ml−1), IL-6 (20 ng ml−1), TPO (50 ng ml−1), Flt3l (50 ng ml−1), SCF (100 ng ml−1) (PeproTech) and 25 mM [U-13C]-glucose (cat. no. CLM-1396–2, Cambridge Isotope Laboratories) at 37 °C for 20 h. After HSC surface staining, cells were washed with ice-cold PBS and resuspended in 0.5 ml ice-cold saline (9 g l−1 NaCl in deionized water). The sorter was configured with a 70-μm nozzle tip and the sheath fluid consisted of 5 g l−1 NaCl in deionized water. A total of 5 × 104 enriched HSCs were sorted directly into 50 μl ice-cold ACN.
Metabolite measurements using LC–MS
Samples were centrifuged at 16,000g for 10 min at 4 °C; 30 μl of supernatant were transferred to MS vials. A quadrupole orbitrap mass spectrometer (Q Exactive, Thermo Fisher Scientific) operating in negative ion mode was coupled to a Vanquish UHPLC system (Thermo Fisher Scientific) with electrospray ionization and used to scan from m/z 70 to 300 at 2 Hz, with a 140,000 resolution. LC separation was achieved on an XBridge BEH Amide Column (2.1 × 150 mm2, 2.5-μm particle size, 130-Å pore size, Waters Corporation) using a gradient of solvent A (95:5 water: ACN with 20 mM ammonium acetate and 20 mM ammonium hydroxide, pH 9.45) and solvent B (ACN). The flow rate was 150 μl min−1. The LC gradient was: 0 min, 85% B; 2 min, 85% B; 3 min, 80% B; 5 min, 80% B; 6 min, 75% B; 7 min, 75% B; 8 min, 70% B; 9 min, 70% B; 10 min, 50% B; 12 min, 50% B; 13 min, 25% B; 16 min, 25% B; 18 min, 0% B; 23 min, 0% B; 24 min, 85% B; and 30 min, 85% B. The autosampler temperature was 5 °C and the injection volume was 15 μl. Data were analyzed using the maven software (build 682; http://maven.princeton.edu/index.php). Natural isotope correction was performed with AccuCor2 R code (https://github.com/wangyujue23/AccuCor2).
ATP quantification
A total of 1 × 104 enriched HSCs were resuspended in 100 μl PBS and equilibrated at room temperature for 30 min before the same volume of Cell Titer-Glo reagent (Promega Corporation) was added. After 10 min incubation at room temperature, the intensity of luminescence was measured using a SpectraiMax i3 Microplate Reader (Molecular Devices).
NAD+ quantification
A total of 1 × 104 enriched HSCs were resuspended in 50 μl PBS; the same volume of NAD/NADH-Glo Detection Reagent (Promega Corporation) was added according to the manufacturer’s instructions. After 60 min incubation at room temperature, the intensity of luminescence was measured using the SpectraiMax i3 Microplate Reader.
Colony-forming assay
A total of 2 × 104 BM cells were used for the colony formation assay in MethoCult GF M3434 (STEMCELL Technologies) according to the manufacturer’s instructions. The colony-forming units were quantified on day 12.
Complete blood count
Peripheral blood was collected via submandibular bleeding into EDTA-treated blood collection tubes. Complete blood count analysis was conducted at the Comparative Pathology Laboratory at the University of California, Davis.
Statistics and reproducibility
The number of mice chosen for each experiment was based on the principle that the minimum number of mice should be used to have sufficient statistical power and was comparable to the published literature for the same assays performed. No statistical method was used to predetermine sample size. Data points were excluded if they were identified as outliers using the Grubbs’ test. No randomization was used to allocate mice to the experimental groups. Analyses of mice and tissue samples were performed by investigators blinded to the treatment or genetic background of the animals. Measurements were taken from distinct samples from different mice. Statistical analysis was performed with Student’s t-tests (in Excel), unless specified otherwise. Data distribution was not tested. Data are presented as the mean while the error bars represent the s.e. In all corresponding figures, *P < 0.05, **P < 0.01, ***P < 0.001, NS P > 0.05. Information about replication is indicated in the figures and figure legends.
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 Material
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.).
Footnotes
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.
Additional information
Extended data is available for this paper at https://doi.org/10.1038/s41563-020-0615-x.
Supplementary information The online version contains supplementary material available at https://doi.org/10.1038/s43587-024-00670-8.
Peer review information Nature Aging thanks Nicola Vannini and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Reprints and permissions information is available at www.nature.com/reprints.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
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
- 1.Linton PJ & Dorshkind K Age-related changes in lymphocyte development and function. Nat. Immunol. 5, 133–139 (2004). [DOI] [PubMed] [Google Scholar]
- 2.Mohrin M et al. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science 347, 1374–1377 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Nakamura-Ishizu A, Ito K & Suda T Hematopoietic stem cell metabolism during development and aging. Dev. Cell 54, 239–255 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Rossi DJ, Jamieson CHM & Weissman IL Stems cells and the pathways to aging and cancer. Cell 132, 681–696 (2008). [DOI] [PubMed] [Google Scholar]
- 5.Mohrin M & Chen D The mitochondrial metabolic checkpoint and aging of hematopoietic stem cells. Curr. Opin. Hematol. 23, 318–324 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wang Y, Barthez M & Chen D Mitochondrial regulation in stem cells. Trends Cell Biol. 10.1016/j.tcb.2023.10.003 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Brown K et al. SIRT3 reverses aging-associated degeneration. Cell Rep. 3, 319–327 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Girotra M et al. Induction of mitochondrial recycling reverts age-associated decline of the hematopoietic and immune systems. Nat. Aging 3, 1057–1066 (2023). [DOI] [PubMed] [Google Scholar]
- 9.Ito K et al. Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science 354, 1156–1160 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Vannini N et al. The NAD-booster nicotinamide riboside potently stimulates hematopoiesis through increased mitochondrial clearance. Cell Stem Cell 24, 405–418 (2019). [DOI] [PubMed] [Google Scholar]
- 12.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Shin J et al. SIRT7 represses Myc activity to suppress ER stress and prevent fatty liver disease. Cell Rep. 5, 654–665 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wang C-L et al. The mitochondrial unfolded protein response regulates hippocampal neural stem cell aging. Cell Metab. 35, 996–1008 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Horenstein AL et al. The circular life of human CD38: from basic science to clinics and back. Molecules 25, 4844 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Howard M et al. Formation and hydrolysis of cyclic ADP-ribose catalyzed by lymphocyte antigen CD38. Science 262, 1056–1059 (1993). [DOI] [PubMed] [Google Scholar]
- 17.Chini EN & De Toledo FGS Nicotinic acid adenine dinucleotide phosphate: a new intracellular second messenger? Am. J. Physiol. Cell Physiol. 282, C1191–C1198 (2002). [DOI] [PubMed] [Google Scholar]
- 18.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). [DOI] [PubMed] [Google Scholar]
- 19.Jin D et al. CD38 is critical for social behaviour by regulating oxytocin secretion. Nature 446, 41–45 (2007). [DOI] [PubMed] [Google Scholar]
- 20.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tarragó MG 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Novelli EM, Ramirez M & Civin CI Biology of CD34+CD38− cells in lymphohematopoiesis. Leuk. Lymphoma 31, 285–293 (1998). [DOI] [PubMed] [Google Scholar]
- 23.Zhao JL 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wilkinson AC, Ishida R, Nakauchi H & Yamazaki S Long-term ex vivo expansion of mouse hematopoietic stem cells. Nat. Protoc. 15, 628–648 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Randall TD, Lund FE, Howard MC & Weissman IL Expression of murine CD38 defines a population of long-term reconstituting hematopoietic stem cells. Blood 87, 4057–4067 (1996). [PubMed] [Google Scholar]
- 27.Suda T, Takubo K & Semenza GL Metabolic regulation of hematopoietic stem cells in the hypoxic niche. Cell Stem Cell 9, 298–310 (2011). [DOI] [PubMed] [Google Scholar]
- 28.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kirichok Y, Krapivinsky G & Clapham DE The mitochondrial calcium uniporter is a highly selective ion channel. Nature 427, 360–364 (2004). [DOI] [PubMed] [Google Scholar]
- 30.Sun X et al. Nicotinamide riboside attenuates age-associated metabolic and functional changes in hematopoietic stem cells. Nat. Commun. 12, 2665 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Covarrubias AJ, 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.He H et al. Aging-induced MCPH1 translocation activates necroptosis and impairs hematopoietic stem cell function. Nat. Aging 4, 510–526 (2024). [DOI] [PubMed] [Google Scholar]
- 33.He H et al. Age-related noncanonical TRMT6-TRMT61A signaling impairs hematopoietic stem cells. Nat. Aging 4, 213–230 (2024). [DOI] [PubMed] [Google Scholar]
- 34.Hong T et al. TET2 modulates spatial relocalization of heterochromatin in aged hematopoietic stem and progenitor cells. Nat. Aging 3, 1387–1400 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wendorff AA et al. Epigenetic reversal of hematopoietic stem cell aging in Phf6-knockout mice. Nat. Aging 2, 1008–1023 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ibneeva L et al. CD38 promotes hematopoietic stem cell dormancy. PLoS Biol. 22, e3002517 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
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.
