Graphical abstract graphic file with name fx1.jpg [71]Open in a new tab Highlights * • Obesity increases CD47 expression while exercise downregulates CD47 expression * • CD47-blocking antibody ameliorates obesity and enhances exercise capacity * • CD47-HSP90α pathway inhibits AMPK activation in skeletal muscle __________________________________________________________________ Su et al. suggest that obesity upregulates CD47 expression while exercise downregulates CD47 expression. Targeting CD47 with a blocking antibody confers multiple metabolic benefits through AMPK signaling in skeletal muscles. These findings reveal a potential therapeutic strategy for obesity-related metabolic disorders. Introduction Obesity results from chronic energy surplus, and prevention or treatment of obesity in humans is elusive. AMP-activated protein kinase (AMPK), an important regulator of energy homeostasis,[72]^1 is a crucial target for treating metabolic diseases such as obesity.[73]^2 A large number of AMPK activators have been developed, like metformin and canagliflozin, which are clinically approved for the treatment of metabolic diseases.[74]^3^,[75]^4 However, most AMPK agonists target multiple tissues to activate AMPK. In some cells or tissues, aberrant activation of AMPK causes detrimental effects on health.[76]^5 For example, prolonged AMPK activation enhances tumor cell viability or induces cardiac hypertrophy.[77]^6 Skeletal muscle accounts for approximately 40% of total body weight, and AMPK activation in skeletal muscle is intrinsically linked to systemic metabolism by increasing glucose uptake and fatty acid (FA) oxidation.[78]^7^,[79]^8^,[80]^9 Consequently, the development of AMPK activators targeting skeletal muscle without cardiac hypertrophy is vital for the treatment of metabolic diseases. CD47 has been widely recognized as a “do-not-eat-me” signal that blocks the phagocytic process of macrophages,[81]^10^,[82]^11 and thus CD47-blocking antibodies are used in clinical trials for the treatment of cancer.[83]^12 A recent study shows that the single-nucleotide polymorphism (SNP) in the CD47 locus is associated with variations in human body shape and composition.[84]^13 It was further reported that systemic genetic deletion of CD47 promotes mitochondrial biogenesis and improves physical performance in mice.[85]^14 However, little is known about the therapeutic role of CD47-blocking antibody in metabolic diseases. Therefore, we evaluated the protective role of CD47-blocking antibody as activator of muscle AMPK against high-fat diet (HFD)-induced metabolic disorders. Furthermore, previous studies have shown that AMPK is a client protein of HSP90α and that reduced levels of HSP90α phosphorylation induce AMPK activation.[86]^15 Therefore, we hypothesized that the CD47-blocking antibody might promote AMPK activation by inhibiting HSP90α phosphorylation. Here, we discovered that obesity upregulates CD47 expression in the skeletal muscle, whereas exercise downregulates its expression in skeletal muscle, indicating CD47 as a potential target for ameliorating metabolic diseases. We found that CD47-blocking antibody promotes AMPK activation in skeletal muscle and prevents metabolic disorders under HFD, conferring metabolic benefits in vivo, including reduced body weight gain, improved body composition, enhanced basal metabolism and exercise capacity, alleviated glucose tolerance, and improved serum lipids and mitochondrial function. Using whole-body or muscle-specific CD47-deficient mice, we similarly confirmed that CD47 loss of function (LOF) exhibited protective effects against HFD-induced metabolic disorders. Our results suggest that CD47-blocking antibody promotes the activation of AMPK by reducing HSP90α phosphorylation in skeletal muscle. Thus, our results reveal the essential role of CD47 signaling in skeletal muscle AMPK activation and explore the potential value of CD47-blocking antibody to treat metabolic diseases. Results Obesity upregulates CD47 expression in skeletal muscle To explore the potential correlation between obesity and CD47 expression, we analyzed the public data from human skeletal muscle (GEO: [87]GSE19420 and GEO: [88]GSE81965); the expression level of CD47 was positively correlated with body mass index (BMI) and homeostasis model assessment insulin resistance (HOMA-IR) ([89]Figures 1A and 1B). Meanwhile, we constructed a mouse model of obesity induced by HFD. Results showed that mRNA and protein expression levels of CD47 were increased in the skeletal muscle tissue of HFD-induced obese mice compared to that in normal diet (ND) controls ([90]Figures 1C and 1D). Figure 1. [91]Figure 1 [92]Open in a new tab Administration of CD47-blocking antibody confers metabolic protective effects in mice under HFD condition (A) Correlation of CD47 expression with BMI in individuals with type 2 diabetes (T2D). (B) Correlation between CD47 expression and HOMA-IR in obese individuals. (C and D) CD47 mRNA and protein expression levels in skeletal muscle of control and obese mice (n = 4). Expression levels quantified, GAPDH as control. (E and F) CD47 mRNA and protein expression in sedentary and exercise-trained mice after 8 weeks (n = 4). Expression quantified, GAPDH as control. (G) Schematic of antibody injection protocol: CD47-blocking antibody (CD47 Ab) or control IgG antibody (Cont) every two days under HFD. (H) Bodyweight changes in mice injected with CD47 Ab or Cont on HFD (n = 6 for Cont and n = 7 for CD47 Ab). (I and J) Body composition measured by NMR in mice (n = 6 for Cont and n = 8 for CD47 Ab). (K) Intraperitoneal glucose tolerance test (IPGTT) and area under the curve (AUC) after 5 weeks HFD administration (n = 6). (L–N) Oxygen consumption, carbon dioxide emissions, and energy expenditure measured using metabolic cage (n = 5 for Cont and n = 6 for CD47 Ab). (O and P) Spontaneous food intake and physical activity (n = 5 for Cont and n = 6 for CD47 Ab). (Q and R) Running distance and duration measured on a treadmill (n = 6). Data are presented as means ± SEM and analyzed by two-tailed Student’s t test (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001; ns, not significant). Furthermore, we analyzed public transcriptomic data (GEO: [93]GSE165630 ) and proteomic data[94]^16 (PRIDE: [95]PXD044445) from the human skeletal muscle of individuals engaged in regular exercise versus sedentary controls. The results indicated that both mRNA and protein levels of CD47 were downregulated in the exercise group ([96]Figures S1A and S1B), suggesting that regular exercise may regulate CD47 expression. To further validate this result, we established an exercise model of mice. Both transcriptional and translational expression levels of CD47 were decreased in the skeletal muscle of the mice with regular exercise compared to those in the sedentary controls ([97]Figures 1E and 1F). Altogether, these results indicate that low expression of CD47 is associated with the beneficial metabolic effects. Administration of CD47-blocking antibody exerts metabolic benefits in mice against HFD To determine whether the functional blockade of CD47 could alleviate HFD-induced metabolic disorders, we subcutaneously injected either a CD47-blocking antibody or an immunoglobulin G (IgG) control antibody into HFD-fed mice ([98]Figure 1G), as described by Kojima et al.[99]^17 Firstly, the mice injected with CD47-blocking antibody displayed a significant decrease in body weight gain on HFD ([100]Figure 1H). Moreover, nuclear magnetic resonance (NMR) results showed that CD47-blocking antibody significantly ameliorated HFD-induced alterations in body composition, as evidenced by a reduction in fat mass and an increase in lean mass ([101]Figures 1I, 1J, [102]S1C, and S1D). Secondly, mice injected with CD47-blocking antibody were protected from HFD-induced glucose intolerance ([103]Figure 1K). To further elucidate the possibilities of body composition improvements, we examined the energy balance in mice injected with CD47-blocking antibody or IgG control. An increase in whole-body basal metabolism ([104]Figures 1L–1N) independent of daily food consumption ([105]Figure 1O) and physical activity ([106]Figure 1P) was observed in mice injected with CD47-blocking antibody. Thirdly, to test whether CD47 function blocking could improve the homeostasis of serum lipids, we measured the serum concentrations of free fatty acids (NEFAs), triglycerides (TGs), total cholesterol (T-CHO), and low-density lipoprotein (LDL-C) in the mice injected with CD47-blocking antibody or IgG control. Results showed that injection with CD47-blocking antibody improved the serum lipid profiles of mice on HFD ([107]Figures S1E–S1H). In parallel with the reduction in serum lipids, we also observed a decrease in lipid accumulation in both skeletal muscle and white adipose tissue (WAT) ([108]Figures S1I–S1L). Most importantly, mice injected with the CD47-blocking antibody exhibited a significant increase in both maximum running distance ([109]Figure 1Q) and duration ([110]Figure 1R) compared to the IgG control group, indicating an improvement in running capacity in the CD47-blocking antibody-treated mice. These findings suggest that CD47-blocking antibody treatment in mice on HFD confers beneficial metabolic effects, including reduced body weight, increased skeletal muscle mass, enhanced basal metabolism, improved glucose homeostasis and serum lipid profiles, reduced lipid droplet accumulation in skeletal muscle, and enhanced exercise capacity. CD47-blocking antibody improves mitochondrial function by promoting AMPK activation in skeletal muscle To elucidate the metabolic benefits of CD47 blockade, we focused on AMPK, a crucial energy sensor that governs metabolic homeostasis.[111]^18 Activation of AMPK in metabolic tissues is known to mitigate obesity, non-alcoholic fatty liver disease (NAFLD), and type 2 diabetes.[112]^7^,[113]^19^,[114]^20^,[115]^21 We hypothesized that CD47 blockade modulates AMPK activation, particularly at Thr172.[116]^22 Indeed, a CD47-blocking antibody activated AMPK in skeletal muscle but not in WAT, brown adipose tissue (BAT), liver, hypothalamus, or kidney ([117]Figure 2A). To explore the potential mechanisms, AMPKγ3 was initially considered due to its selective expression in skeletal muscle,[118]^23 which aligns with our findings ([119]Figure S2A). To further investigate the possible role of the AMPKγ3 subunit in CD47-blocking antibody-induced AMPK activation, we knocked down the γ3 subunit in differentiated myotubes and treated them with a CD47-blocking antibody. As shown in [120]Figures S2B and S2C, the loss of γ3 expression abolished AMPK activation induced by the CD47-blocking antibody, indicating that the γ3 subunit plays an important role in CD47-blocking antibody-mediated AMPK activation in skeletal muscle. The AMPK activity in the skeletal muscle mainly originates from the AMPKγ3 complex and the AMPKγ1 complex.[121]^7^,[122]^24 To further evaluate the relative contributions of γ3 and γ1 subunits to AMPK activation, we measured the activities of AMPK complexes containing γ3 or γ1 in the muscle of mice treated with a CD47-blocking antibody in vivo, following the protocol outlined in [123]Figure S2D.[124]^23 The results showed that the AMPKγ3 complex was activated to a greater extent than the AMPKγ1 complex ([125]Figure S2E). Figure 2. [126]Figure 2 [127]Open in a new tab CD47-blocking antibody promotes skeletal muscle AMPK activation and improves mitochondrial function (A) AMPK phosphorylation in skeletal muscle, WAT, BAT, liver, hypothalamus (Hypo), and kidney (n = 6). (B) ACC phosphorylation in skeletal muscle after antibody injection. GAPDH as control. (C) PGC-1α protein levels in skeletal muscle post-injection. GAPDH as control. (D) Western blot analysis of AMPK, ACC phosphorylation, and PGC-1α in skeletal muscle of mice injected with CD47 Ab (20 mg/kg), Cont (20 mg/kg), and AMPK inhibitor (Compound C, 10 mg/kg) (n = 3). (E and F) AMPK phosphorylation and cytochrome c (cyto c) protein expression in myotubes after antibody treatment. GAPDH as control. (G) Seahorse mitochondrial stress test in myotubes treated with CD47 Ab. (H–K) AMPK, ACC phosphorylation, and PGC-1α expression in myotubes with Cd47 knockdown or overexpression. (L) Seahorse mitochondrial stress test in myotubes with Cd47 knockdown. Data are presented as means ± SEM and analyzed by two-tailed Student’s t test (A and G) and two-way repeated measures (RM) ANOVA (D and L). ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001; ns, not significant. AMPK activation in skeletal muscle promotes FA oxidation[128]^24 and mitochondrial respiration.[129]^25 Accordingly, we examined acetyl-coenzyme A carboxylase (ACC), a key FA-oxidation regulator that, when phosphorylated by AMPK, increases fat oxidation.[130]^26 In mice treated with a CD47-blocking antibody, ACC phosphorylation (pSer79) was elevated in skeletal muscle ([131]Figure 2B). Because peroxisome proliferator-activated receptor-gamma co-activator-1 alpha (PGC-1α) enhances mitochondrial respiration,[132]^27 we next observed that CD47 blockade also increased PGC-1α expression ([133]Figure 2C). In vivo administration of the AMPK inhibitor Compound C suppressed the antibody-induced phosphorylation of AMPK and ACC, as well as the upregulation of PGC-1α ([134]Figure 2D), confirming an AMPK-dependent mechanism. Consistently, CD47 blockade in vitro activated AMPK in myotubes and raised cytochrome c (cyto c) levels, improving maximal respiration and ATP production ([135]Figures 2E–2G). These findings suggest that, under HFD conditions, CD47 blockade augments skeletal muscle mitochondrial energetics. To further examine the role of CD47 in regulating AMPK activation, we knocked down or overexpressed Cd47 in myotubes ([136]Figures S3A and S3B). Cd47 deficiency markedly increased AMPK phosphorylation and downstream ACC (pSer79), whereas Cd47 overexpression suppressed both ([137]Figures 2H and 2I), confirming that CD47 negatively regulates the AMPK/ACC axis. Likewise, PGC-1α was upregulated in Cd47 knockdown myotubes but reduced upon overexpression ([138]Figures 2J and 2K), highlighting the role of CD47 in regulating mitochondrial function. Seahorse analysis showed that Cd47 knockdown enhanced maximal respiration and ATP production ([139]Figure 2L). Beyond AMPK, the mitochondrial energetics regulator SIRT1[140]^28^,[141]^29 was also elevated in Cd47-deficient myotubes ([142]Figure S3C) and in skeletal muscle of CD47^−/− mice ([143]Figure S3D). Because both AMPK and SIRT1 converge on PGC-1α to bolster mitochondrial function,[144]^30 these results underscore coordinated regulation by CD47 LOF in skeletal muscle. CD47 LOF in skeletal muscle promotes AMPK activation Next, we generated CD47 whole-body knockout (CD47^−/−) by the CRISPR/Cas9 system ([145]Figures 3A, [146]S4A, and S4B). Firstly, the CD47^−/− mice displayed a significant decrease in body weight gain ([147]Figures 3B and [148]S4C). Further studies revealed a significant improvement in body composition in CD47^−/− mice, as evidenced by a decrease in fat mass and an increase in lean mass ([149]Figures S4D and S4E). Secondly, an improvement in glucose tolerance was observed in CD47^−/− mice ([150]Figure 3C). In addition, CD47 LOF significantly improved serum lipid profiles ([151]Figures S4F–S4J). Along with the reduction in serum lipids, we observed a decrease in lipid accumulation in both skeletal muscle and WAT ([152]Figures S4K and S4L). Finally, the maximal running distance ([153]Figure S4M) and duration ([154]Figure S4N) were significantly greater in CD47^−/− mice compared to wild-type (WT) mice. Figure 3. [155]Figure 3 [156]Open in a new tab CD47 LOF in skeletal muscle promotes AMPK activation (A) Schematic of CD47-knockout (CD47^−/−) mouse model. (B) Growth curves of WT and CD47^−/− mice (n = 6). (C) IPGTT and AUC in WT and CD47^−/− mice (n = 6). (D and E) Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of differentially expressed genes in skeletal muscle of CD47^−/− and exercise mice. (F) AMPK and ACC phosphorylation in skeletal muscle of WT and CD47^−/− mice. (G) PGC-1α, cyto c, and oxidase (COXIV) expression levels in skeletal muscle of WT and CD47^−/− mice. (H and I) ATPase and CS activities in skeletal muscle of WT and CD47^−/− mice (n = 7). (J) Schematic of MCK-CD47 mouse model. (K) Growth curves of MCK-CD47 mice on HFD (n = 10 for Con and n = 6 for MCK-CD47). (L) IPGTT and AUC in MCK-CD47 mice after 3 months of HFD (n = 10 for Con and n = 6 for MCK-CD47). (M) AMPK phosphorylation and PGC-1α expression in skeletal muscle of MCK-CD47 mice. (N) CS activity in skeletal muscle of MCK-CD47 mice (n = 11 for Con and n = 7 for MCK-CD47). (O) Schematic of HSA-CD47 mouse model. (P and Q) Growth curves and IPGTT of HSA-CD47 mice on HFD (n = 8 for Con and n = 6 for HSA-CD47). (R and S) Running distance and duration of HSA-CD47 mice on motor treadmill (n = 9 for Con and n = 7 for HSA-CD47). (T) CS activity in skeletal muscle of HSA-CD47 mice (n = 9 for Con and n = 7 for HSA-CD47). (U) AMPK phosphorylation and PGC-1α expression in skeletal muscle of HSA-CD47 mice. (V) Western blot analysis of CD47 and AMPK phosphorylation in skeletal muscle of mice injected with AAV expressing GFP or CD47. Data are presented as means ± SEM and analyzed by two-tailed Student’s t test (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001; ns, not significant). To further investigate how CD47 LOF exerts beneficial metabolic effects in vivo, RNA sequencing (RNA-seq) was performed in the skeletal muscle of the CD47^−/− mice, WT mice with regular exercise (Ex), and sedentary WT controls (Con). Notably, genes associated with the AMPK pathway were highly enriched in the skeletal muscle of both CD47^−/− mice and Ex mice compared to the Con mice ([157]Figures 3D and 3E). Consistent with findings from Cd47-deficient myotubes, the phosphorylation levels of AMPK (pThr172) and ACC (pSer79) were increased in the skeletal muscle of CD47^−/− mice compared to littermate WT controls ([158]Figure 3F). In addition, PGC-1α ([159]Figure 3G) level was also increased in the skeletal muscle of CD47^−/− mice. Meanwhile, mitochondrial enzymes including cyto c, oxidase (COXIV) protein level ([160]Figure 3G), and adenosine triphosphatase (ATPase) ([161]Figure 3H), as well as citrate synthase (CS) ([162]Figure 3I), were increased in skeletal muscle of CD47^−/− mice. These results are consistent with those in CD47-blocking antibody-injected mice. Mitochondria metabolism and function are tightly regulated by protein acetylation.[163]^31 Analysis of the mitochondrial acetylome revealed that over 60% of mitochondrial proteins contain acetylation sites, with most involved in energy metabolism, particularly FA metabolism, the tricarboxylic acid (TCA) cycle, the electron transport chain, and oxidative phosphorylation (OXPHOS).[164]^32 SIRT3 is one of the sirtuins localized in mitochondria that deacetylates key mitochondrial proteins essential for metabolism and oxidative homeostasis.[165]^33^,[166]^34^,[167]^35 Our findings indicated that Cd47 gene deletion was associated with a significant increase in SIRT3 expression levels ([168]Figure S5A). Given the critical role of SIRT3 in the regulation of mitochondrial metabolism,[169]^31 we subsequently isolated mitochondria from the skeletal muscle of CD47^−/− mice and investigated the changes in mitochondrial protein acetylation levels. The results showed that CD47 deletion led to a significant reduction in protein acetylation levels of mitochondria ([170]Figure S5B). To further assess protein acetylation, we performed a proteomic analysis that identified 1,530 acetylation sites on 410 skeletal muscle proteins. Of these, 53 acetylation sites were significantly altered in the skeletal muscle tissues of CD47^−/− mice ([171]Table S1). These altered sites include several deacetylated mitochondrial proteins, such as Iba57, ldh3a, and Slc25a12, with ldh3a playing a key role in the TCA pathway ([172]Figure S5C). To investigate the muscle-specific role of CD47 in regulating AMPK activation, we generated muscle-specific CD47-knockout (MCK-CD47) mice by crossing Cd47^flox/flox mice with MCK-Cre mice ([173]Figure 3J). After 20 weeks on an HFD, MCK-CD47 mice exhibited significantly reduced weight gain compared with littermate controls ([174]Figures 3K and [175]S6A), along with lower random blood glucose ([176]Figure S6B), improved glucose tolerance ([177]Figure 3L) and insulin sensitivity ([178]Figure S6C), and reduced lipid accumulation in WAT and skeletal muscle ([179]Figures S6D and S6E). Consistent with results from CD47-blocking antibody-treated mice, MCK-CD47 mice displayed enhanced AMPK activation in skeletal muscle ([180]Figure 3M). Building on the improved mitochondrial energetics observed under CD47 blockade, we found that MCK-CD47 mice showed elevated PGC-1α expression ([181]Figure 3M) and CS activity ([182]Figure 3N). To extend these findings, we generated human α-skeletal actin (HSA)-CD47 mice (Cd47^flox/flox, Hsa-MerCreMer) with CD47 selectively deleted in mature muscle cells ([183]Figure 3O). Similarly, HFD-fed HSA-CD47 mice exhibited significantly lower body weight ([184]Figure 3P), improved glucose tolerance ([185]Figure 3Q), and greater running distance and duration ([186]Figures 3R and 3S). Their skeletal muscle displayed increased CS activity ([187]Figure 3T), AMPK activation, and PGC-1α expression ([188]Figure 3U). Finally, to confirm that CD47 indeed regulates AMPK activity, we administered an adeno-associated virus (AAV) overexpressing CD47 intramuscularly, which significantly suppressed AMPK activation ([189]Figure 3V). Together, these data demonstrate that CD47 directly modulates AMPK activation in skeletal muscle. CD47-blocking antibody promotes AMPK activation by reducing phosphorylation of HSP90α To gain insight into the mechanism of AMPK activation by CD47 LOF, we constructed a cell line stably expressing FLAG-tagged CD47 in myotubes and then performed co-immunoprecipitation (coIP) and mass spectrometry (MS) analysis to identify proteins that may interact with CD47 ([190]Figure 4A). Several proteins related to metabolism were identified by MS analyses, including HSP90α, GNB2, GNB4, SLC25A4, UGCG, HSPA8, PKM, and UBC ([191]Figures S7A and S7B). The direct interaction between CD47 and HSP90α was further verified by coIP assay in 293T cells ([192]Figure S7C) and differentiated myotubes ([193]Figure 4B). The results indicated that CD47 interacted with HSP90α. Previous studies have demonstrated that HSP90α interacts with AMPK[194]^36 and that decreased phosphorylation of HSP90α leads to increased activation of AMPK.[195]^15 Furthermore, DNA-dependent protein kinase (DNA-PK), a serine/threonine kinase, has been implicated in the phosphorylation of HSP90α in several studies.[196]^15^,[197]^37^,[198]^38 Here, we speculated that CD47-blocking antibody inhibits HSP90α phosphorylation via DNA-PK, which in turn promotes AMPK activation. To test this, we investigated the effects of Cd47 knockdown, overexpression, and CD47 functional blockade on DNA-PK activity and phosphorylation of its downstream target, HSP90α. DNA-PK activity (p-DNA-PK) was assessed by monitoring the autophosphorylation at S2056.[199]^39 The results revealed that Cd47 deletion reduced the phosphorylation of both DNA-PK and HSP90α ([200]Figure 4C), while Cd47 overexpression had the opposite effect, enhancing their phosphorylation ([201]Figure 4D). To determine whether p-DNA-PK mediates CD47-induced HSP90α phosphorylation, myotubes overexpressing Cd47 were treated with a DNA-PK inhibitor. The results showed that DNA-PK inhibition abolished the Cd47 overexpression-induced phosphorylation of HSP90α ([202]Figure 4E), suggesting that activated DNA-PK is essential for CD47 regulation of HSP90α phosphorylation. Consistently, the phosphorylation levels of both DNA-PK and HSP90α were also reduced in myotubes treated with a CD47-blocking antibody ([203]Figure 4F). Collectively, these findings indicate that the CD47-blocking antibody reduces HSP90α phosphorylation via inhibition of DNA-PK. Figure 4. [204]Figure 4 [205]Open in a new tab CD47-blocking antibody promotes AMPK activation by reducing phosphorylation of HSP90α (A) Strategy for analyzing CD47-interacting proteins using liquid chromatography-tandem mass spectrometry (LC-MS/MS). (B) Endogenous interactions between CD47 and HSP90α detected in myotubes by western blotting. (C and D) HSP90α and DNA-PK phosphorylation in Cd47 knockdown or overexpressing myotubes. (E) DNA-PK inhibitor treatment (10 μM) effects on HSP90α and DNA-PK phosphorylation in Cd47-overexpressing myotubes. (F) HSP90α and DNA-PK phosphorylation in myotubes treated with CD47-blocking antibody. (G) HSP90α phosphorylation and AMPK activation after treatment with tanespimycin (17-AAG). (H) AMPK phosphorylation in Cd47-overexpressing myotubes treated with 17-AAG. (I) AMPK phosphorylation after lentiviral treatment with mutated HSP90α phosphorylation sites. (J) The inhibitory effect of Cd47 overexpression on AMPK activation was blocked by mutated HSP90α phosphorylation sites. (K) Schematic: CD47-blocking antibody reduces HSP90α phosphorylation, promoting AMPK activation. Data are presented as means ± SEM and analyzed by two-way RM ANOVA (C, right) and Student’s t test (D, right). ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001; ns, not significant. To verify whether CD47 inhibits AMPK activation by increasing HSP90α phosphorylation, an inhibitor of HSP90α, tanespimycin (17-AAG),[206]^40 was added into Cd47-overexpressing myotubes. Our results showed that 17-AAG reduced the phosphorylation level of HSP90α and increased the activation of AMPK ([207]Figure 4G); meanwhile, the inhibitory effect of Cd47 overexpression on AMPK activation was also effectively blocked by 17-AAG ([208]Figure 4H). To further demonstrate that CD47 inhibits AMPK activation by increasing HSP90α phosphorylation levels, lentiviruses with normal and mutant HSP90α (inhibiting phosphorylation) sites were utilized to infect Cd47-overexpressing myotubes. The results showed that myotube cells infected with the mutant HSP90α could relieve the inhibition of AMPK activation by Cd47 overexpression ([209]Figure 4I). In sum, these results suggest that CD47 promotes HSP90α phosphorylation to inhibit AMPK activation ([210]Figure 4J). Interestingly, 17-AAG was administered intraperitoneally in mice on HFD, and, through long-term monitoring, the results showed that the inhibitor effectively alleviated HFD-induced obesity ([211]Figure S7D) and glucose homeostasis ([212]Figures S7E and S7F), promoted AMPK activation, and elevated PGC-1α levels in skeletal muscle tissue ([213]Figure S7G), consistent with the phenotype observed in mice injected with CD47-blocking antibody. In conclusion, the CD47-blocking antibody promotes AMPK activation by reducing phosphorylation of HSP90α ([214]Figure 4K). In order to clinically translate CD47 antibodies, it is essential to evaluate their side effects in vivo. One of the most significant questions is whether their long-term application could lead to cardiac hypertrophy caused by persistent AMPK activation. In this study, our results revealed no significant difference in heart sizes ([215]Figures S8A and S8B); heart weight/body weight ratios ([216]Figure S8C); cardiomyocyte cross-sectional area, as assessed by wheat germ agglutinin (WGA) staining ([217]Figure S8D); or cardiac fibrosis, as assessed by Masson’s trichrome staining ([218]Figure S8E) between CD47^−/− and WT mice. Furthermore, no differences were observed in left ventricular diastolic and systolic posterior wall thickness (LVPW:d, LVPW:s) ([219]Figures S8F and S8G) in CD47-blocking antibody-injected mice compared to control IgG antibody-injected mice. In brief, our results did not reveal any significant changes in cardiac morphology and fibrosis following CD47 deficiency or blockade. Notably, CD47-blocking antibody treatment under HFD conditions significantly attenuated the reduction in ejection fraction and fractional shortening ([220]Figures S8H and S8I), suggesting an improvement in cardiac function. Collectively, these findings demonstrate that long-term treatment with CD47-blocking antibodies does not induce cardiac hypertrophy. Previous studies have shown that both CD47-blocking antibodies and Cd47 gene knockout effectively mitigate isoproterenol-induced cardiac hypertrophy.[221]^41^,[222]^42 Other research suggests that CD47 expression is upregulated during cardiac hypertrophy.[223]^43^,[224]^44 In conjunction with our findings, these observations strengthen the hypothesis that CD47 deficiency or blockade may play a pivotal role in preserving cardiac health under both pathological and physiological conditions. Discussion In the current study, we demonstrated a direct causal link between CD47 LOF and AMPK activation in three CD47-knockout mouse models and further presented a rationale for the therapeutic potential of CD47-blocking antibodies in metabolic diseases by targeting skeletal muscle AMPK activation. CD47 pharmacological intervention can offer a promising approach to alleviate HFD-induced metabolic disorders, conferring metabolic benefits. CD47-blocking antibodies have been used in clinical trials for the treatment of cancer.[225]^45 Here, our results revealed that the administration of the CD47-blocking antibody exerts metabolic benefits against HFD, including reduced body weight gain, improved body composition, enhanced basal metabolism and exercise capacity, alleviated glucose tolerance, and improved serum lipids and mitochondrial function. Notably, most weight-loss drugs tend to reduce muscle mass while losing body weight.[226]^46 Here, we found that, in addition to its significant therapeutic effect on weight loss, CD47-blocking antibody also preserves muscle mass while reducing fat mass. This may be attributed to the essential role of CD47 signaling in the expansion of muscle stem cells (MuSCs).[227]^47 The discovery has important clinical implications. (1) CD47-blocking antibody is an important candidate for the development of weight-loss drugs. (2) It provides a potential strategy for expanding endogenous MuSCs in vivo for therapeutic purposes. In addition, whether CD47-blocking antibody treatment has curative effects on skeletal muscle dysfunction due to other pathological conditions, such as muscular dystrophy diseases, is unclear. Therefore, the applications of CD47-blocking antibodies should be expanded in future studies. CD47 plays distinct roles in different cells.[228]^48^,[229]^49^,[230]^50^,[231]^51 In detail, as a transmembrane protein, CD47 is a counter-receptor for signal-regulated protein-alpha (SIRPα). The CD47-SIRPα axis is crucial in delivering the “do-not-eat-me” signal, protecting cells from phagocytosis by macrophages.[232]^52 Thrombospondin-1 (TSP1) is the other receptor for CD47, and the TSP1-CD47 axis is necessary for MuSCs proliferation under injury.[233]^47 A recent study reveals that TSP1/CD47 paracrine signaling inhibits the proliferation of aged MuSCs, and blocking the TSP1/CD47 signaling restores the proliferative capacity of MuSCs.[234]^53 However, due to the lack of a cytoplasmic domain of CD47, these studies mainly examined the role of CD47 in extracellular signaling with a limited understanding of intracellular downstream signaling.[235]^54 Here, our study elucidates the intracellular pathway linking CD47 to AMPK activation and highlights a therapeutic potential for CD47-blocking antibodies. Limitations of the study In conclusion, our study highlights that the CD47-blocking antibody confers metabolic protective effects against obesity via restoration of AMPK activity in skeletal muscles. We show that CD47 inhibits AMPK activity through the DNA-PK-HSP90α pathway, which aligns with previous reports on the metabolic regulatory function of DNA-PK.[236]^15 Future studies should investigate whether this signaling mechanism is also operative in other cell types or under different pathological conditions, which will further elucidate the therapeutic efficacy of CD47-blocking antibody and broaden its potential clinical applications. We observe an interaction between CD47 and HSP90α, with CD47 promoting HSP90α phosphorylation. These results infer that CD47 may facilitate HSP90α phosphorylation by stabilizing the interaction between DNA-PK and HSP90α. However, the precise mechanism by which CD47 modulates the DNA-PK-HSP90α-AMPK axis—particularly whether this occurs through receptor-mediated signaling or direct interaction—requires further investigation. Additionally, the potential involvement of the cellular AMP/ATP ratio or Ca^2+-CaMKK2 signaling in the downstream effects of CD47 and how these signaling pathways interact to regulate AMPK activity also warrant future investigation. Resource availability Lead contact For additional information or material requests, please contact the corresponding author, Weida Li (liweida@tongji.edu.cn), who will coordinate all related inquiries and requests. Materials availability All materials and reagents generated in this study are available from the corresponding author upon execution of an appropriate materials transfer agreement. Data and code availability * • All data associated with this study are included in the manuscript and its [237]supplemental information. * • The RNA-seq raw data and processed count matrices generated in this study have been deposited in the NCBI Sequence Read Archive (SRA) under BioProject accession SRA: PRJNA1224594, and the proteomics dataset is available via the PRIDE database under accession PRIDE: [238]PXD061117. Published transcriptomic data analyzed in this work were obtained from GEO: [239]GSE19420 , GEO: [240]GSE81965, GEO: [241]GSE165630, and proteomic data were sourced from PRIDE: [242]PXD044445. * • This paper does not report original code. * • Any additional information required to reanalyze the data reported in this paper is available from the [243]lead contact upon request. Acknowledgments