AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (7.2 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

A novel multifunctional radioprotective strategy using P7C3 as a countermeasure against ionizing radiation-induced bone loss

Fei Wei1Zewen Kelvin Tuong2,3Mahmoud Omer1Christopher Ngo1Jackson Asiatico4Michael Kinzel4Abinaya Sindu Pugazhendhi1Annette R. Khaled5Ranajay Ghosh4Melanie Coathup1( )
Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
Show Author Information

Abstract

Radiotherapy is a critical component of cancer care but can cause osteoporosis and pathological insufficiency fractures in surrounding and otherwise healthy bone. Presently, no effective countermeasure exists, and ionizing radiation-induced bone damage continues to be a substantial source of pain and morbidity. The purpose of this study was to investigate a small molecule aminopropyl carbazole named P7C3 as a novel radioprotective strategy. Our studies revealed that P7C3 repressed ionizing radiation (IR)-induced osteoclastic activity, inhibited adipogenesis, and promoted osteoblastogenesis and mineral deposition in vitro. We also demonstrated that rodents exposed to clinically equivalent hypofractionated levels of IR in vivo develop weakened, osteoporotic bone. However, the administration of P7C3 significantly inhibited osteoclastic activity, lipid formation and bone marrow adiposity and mitigated tissue loss such that bone maintained its area, architecture, and mechanical strength. Our findings revealed significant enhancement of cellular macromolecule metabolic processes, myeloid cell differentiation, and the proteins LRP-4, TAGLN, ILK, and Tollip, with downregulation of GDF-3, SH2B1, and CD200. These proteins are key in favoring osteoblast over adipogenic progenitor differentiation, cell matrix interactions, and shape and motility, facilitating inflammatory resolution, and suppressing osteoclastogenesis, potentially via Wnt/β-catenin signaling. A concern was whether P7C3 afforded similar protection to cancer cells. Preliminarily, and remarkably, at the same protective P7C3 dose, a significant reduction in triple-negative breast cancer and osteosarcoma cell metabolic activity was found in vitro. Together, these results indicate that P7C3 is a previously undiscovered key regulator of adipo-osteogenic progenitor lineage commitment and may serve as a novel multifunctional therapeutic strategy, leaving IR an effective clinical tool while diminishing the risk of adverse post-IR complications. Our data uncover a new approach for the prevention of radiation-induced bone damage, and further work is needed to investigate its ability to selectively drive cancer cell death.

References

1

Bayat Mokhtari, R. et al. Combination therapy in combating cancer. Oncotarget 8, 38022–38043 (2017).

2
Berkey, F. J. Managing the adverse effects of radiation therapy. Am. Fam. Physician 82, 381–388 (2010). 394.
3

Jaffray, D. A. et al. Global task force on radiotherapy for cancer control. Lancet Oncol. 16, 1144–1146 (2015).

4

Jarosz-Biej, M., Smolarczyk, R., Cichoń, T. & Kułach, N. Tumor microenvironment as a “game changer” in cancer radiotherapy. Int. J. Mol. Sci. 20, 3212 (2019).

5

Curi, M. M. et al. Histopathologic and histomorphometric analysis of irradiation injury in bone and the surrounding soft tissues of the jaws. J. Oral. Maxillofac. Surg. 74, 190–199 (2016).

6

Daniel, M., Luby, A. O., Buchman, L. & Buchman, S. R. Overcoming nuclear winter: the cutting-edge science of bone healing and regeneration in irradiated fields. Plast. Reconstr. Surg. Glob. Open 9, e3605 (2021).

7

Oh, D. & Huh, S. J. Insufficiency fracture after radiation therapy. Radiat. Oncol. J. 32, 213 (2014).

8

Sparks, R. B., Crowe, E. A., Wong, F. C., Toohey, R. E. & Siegel, J. A. Radiation dose distributions in normal tissue adjacent to tumors containing (131)I or (90)Y: the potential for toxicity. J. Nucl. Med. 43, 1110–1114 (2002).

9

Willey, J. S., Lloyd, S. A. J., Nelson, G. A. & Bateman, T. A. Ionizing radiation and bone loss: space exploration and clinical therapy applications. Clin. Rev. Bone Min. Metab. 9, 54–62 (2011).

10

Soares, C. B. G. et al. Pathological fracture after radiotherapy: systematic review of literature. Rev. Assoc. Med. Bras. 65, 902–908 (2019).

11

Donaubauer, A.-J., et al. The influence of radiation on bone and bone cells—differential effects on osteoclasts and osteoblasts. Int. J. Mol. Sci. 21, 6377 (2020).

12

Kim, H. J., et al. Fractures of the sacrum after chemoradiation for rectal carcinoma: incidence, risk factors, and radiographic evaluation. Int. J. Radiat. Oncol. 84, 694–699 (2012).

13
Aoki, M. et al. Riation-induced rib fracture after stereotactic body radiotherapy with a total dose of 54-56 Gy given in 9-7 fractions for patients with peripheral lung tumor: impact of maximum dose and fraction size.Radiat. Oncol. 10, 99, https://doi.org/10.1186/s13014-015-0406-8 (2015).
14

Blomlie, V., et al. Incidence of radiation-induced insufficiency fractures of the female pelvis: evaluation with MR imaging. Am. J. Roentgenol. 167, 1205–1210 (1996).

15

Gu, J. et al. Effect of amifostine in head and neck cancer patients treated with radiotherapy: a systematic review and meta-analysis based on randomized controlled trials. PLoS One 9, e95968 (2014).

16

Capizzi, R. L. The preclinical basis for broad-spectrum selective cytoprotection of normal tissues from cytotoxic therapies by amifostine. Semin. Oncol. 26, 3–21 (1999).

17

Singh, V. K. & Seed, T. M. The efficacy and safety of amifostine for the acute radiation syndrome. Expert Opin. Drug Saf. 18, 1077–1090 (2019).

18

Yasueda, A., Urushima, H. & Ito, T. Efficacy and interaction of antioxidant supplements as adjuvant therapy in cancer treatment: a systematic review. Integr. Cancer Ther. 15, 17–39 (2016).

19

Shirazi, A., GhobadiI, G. & Ghazi-Khansari, M. A radiobiological review on melatonin: a novel radioprotector. J. Radiat. Res. 48, 263–272 (2007).

20

Wissing, M. D. Chemotherapy- and irradiation-induced bone loss in adults with solid tumors. Curr. Osteoporos. Rep. 13, 140–145 (2015).

21

Sung, H. et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021).

22

American Cancer Society. Global Cancer Facts & Figures 4th edn. Atlanta: American Cancer Society. 2018

23

Bharath, A. K. & Turner, R. J. Impact of climate change on skin cancer. J. R. Soc. Med. 102, 215–218 (2009).

24

Wild, C. P. The global cancer burden: necessity is the mother of prevention. Nat. Rev. Cancer 19, 123–124 (2019).

25

Lustberg, M. B., Reinbolt, R. E. & Shapiro, C. L. Bone health in adult cancer survivorship. J. Clin. Oncol. 30, 3665–3674 (2012).

26

Coleman, R., Body, J. J., Aapro, M., Hadji, P. & Herrstedt, J. Bone health in cancer patients: ESMO clinical practice guidelines. Ann. Oncol. 25, iii124–iii137 (2014).

27

Szymczyk, K. H., Shapiro, I. M. & Adams, C. S. Ionizing radiation sensitizes bone cells to apoptosis. Bone 34, 148–156 (2004).

28

Fekete, N. et al. Effect of high-dose irradiation on human bone-marrow-derived mesenchymal stromal cells. Tissue Eng. Part C. Methods 21, 112–122 (2015).

29

Gal, T. J., Munoz-Antonia, T., Muro-Cacho, C. A. & Klotch, D. W. Radiation effects on osteoblasts in vitro: a potential role in osteoradionecrosis. Arch. Otolaryngol. Head. Neck Surg. 126, 1124–1128 (2000).

30

Dong, J. et al. The combined effects of simulated microgravity and X-ray radiation on MC3T3-E1 cells and rat femurs. npj Microgravity 7, 3 (2021).

31

Kondo, H. et al. Oxidative stress and gamma radiation-induced cancellous bone loss with musculoskeletal disuse. J. Appl. Physiol. 108, 152–161 (2010).

32

Wei, F. et al. A novel approach for the prevention of ionizing radiation-induced bone loss using a designer multifunctional cerium oxide nanozyme. Bioact. Mater. 21, 547–565 (2023).

33

Boyce, B. F. & Xing, L. The RANKL/RANK/OPG pathway. Curr. Osteoporos. Rep. 5, 98–104 (2007).

34

Alwood, J. S. et al. Ionizing radiation stimulates expression of pro-osteoclastogenic genes in marrow and skeletal tissue. J. Inter. Cytokine Res. 35, 480–487 (2015).

35

Amjad, S. et al. Role of NAD+ in regulating cellular and metabolic signaling pathways. Mol. Metab. 49, 101195 (2021).

36

Covarrubias, A. J., Perrone, R., Grozio, A. & Verdin, E. NAD+ metabolism and its roles in cellular processes during ageing. Nat. Rev. Mol. Cell Biol. 22, 119–141 (2021).

37

Verdin, E. NAD+ in aging, metabolism, and neurodegeneration. Science (80-) 350, 1208–1213 (2015).

38

Hong, S. M. et al. Increased nicotinamide adenine dinucleotide pool promotes colon cancer progression by suppressing reactive oxygen species level. Cancer Sci. 110, 629–638 (2019).

39

Schultz, M. B. & Sinclair, D. A. Why NAD(+) declines during aging: it’s destroyed. Cell Metab. 23, 965–966 (2016).

40

Kim, H.-N. et al. A decrease in NAD+ contributes to the loss of osteoprogenitors and bone mass with aging. NPJ Aging Mech. Dis. 7, 8 (2021).

41

Lewis, J. E. et al. Targeting NAD+ metabolism to enhance radiation therapy responses. Semin. Radiat. Oncol. 29, 6–15 (2019).

42

Alano, C. C. et al. NAD+ depletion is necessary and sufficient forPoly(ADP-Ribose) polymerase-1-mediated neuronal death. J. Neurosci. 30, 2967–2978 (2010).

43

Wang, G. et al. P7C3 neuroprotective chemicals function by activating the rate-limiting enzyme in NAD salvage. Cell 158, 1324–1334 (2014).

44

Zhang, T. et al. Enzymes in the NAD+ salvage pathway regulate SIRT1 activity at target gene promoters. J. Biol. Chem. 284, 20408–20417 (2009).

45

Bauman, M. D. et al. Neuroprotective efficacy of P7C3 compounds in primate hippocampus. Transl. Psychiatry 8, 202 (2018).

46

Kim, S. P. et al. Lrp4 expression by adipocytes and osteoblasts differentially impacts sclerostin’s endocrine effects on body composition and glucose metabolism. J. Biol. Chem. 294, 6899–6911 (2019).

47

Elsafadi, M. et al. Transgelin is a TGFβ-inducible gene that regulates osteoblastic and adipogenic differentiation of human skeletal stem cells through actin cytoskeleston organization. Cell Death Dis. 7, e2321–e2321 (2016).

48

Shen, J. J. et al. Deficiency of growth differentiation factor 3 protects against diet-induced obesity by selectively acting on white adipose. Mol. Endocrinol. 23, 113–123 (2009).

49

Rui, L. SH2B1 regulation of energy balance, body weight, and glucose metabolism. World J. Diabetes 5, 511 (2014).

50

Ong, W. K. et al. Identification of specific cell-surface markers of adipose-derived stem cells from subcutaneous and visceral fat depots. Stem Cell Rep. 2, 171–179 (2014).

51

Wu, C. & Dedhar, S. Integrin-linked kinase (ILK) and its interactors. J. Cell Biol. 155, 505–510 (2001).

52

Schaunaman, N., Dimasuay, K. G., Kraft, M. & Chu, H. W. Tollip interaction with STAT3: a novel mechanism to regulate human airway epithelial responses to type 2 cytokines. Respir. Res. 23, 31 (2022).

53

Aitken, C. J. et al. Regulation of human osteoclast differentiation by thioredoxin binding protein-2 and redox-sensitive signaling. J. Bone Min. Res. 19, 2057–2064 (2004).

54

Vignery, A. Osteoclasts and giant cells: macrophage-macrophage fusion mechanism. Int. J. Exp. Pathol. 81, 291–304 (2004).

55

Wei, F. et al. Cerium oxide nanoparticles protect against irradiation-induced cellular damage while augmenting osteogenesis. Mater Sci. Eng. C. 126, 112145 (2021).

56

Nassour, J. et al. Defective DNA single-strand break repair is responsible for senescence and neoplastic escape of epithelial cells. Nat. Commun. 7, 10399 (2016).

57

Murray Brunt, A. et al. Hypofractionated breast radiotherapy for 1 week versus 3 weeks (FAST-Forward): 5-year efficacy and late normal tissue effects results from a multicentre, non-inferiority, randomised, phase 3 trial. Lancet 395, 1613–1626 (2020).

58

Murthy, V. et al. Elective nodal dose of 60 Gy or 50 Gy in head and neck cancers: a matched pair analysis of outcomes and toxicity. Adv. Radiat. Oncol. 2, 339–345 (2017).

59

Hegde, J. V. et al. Head and neck cancer reirradiation with interstitial high-dose-rate brachytherapy. Head. Neck 40, 1524–1533 (2018).

60

Donneys, A. et al. Amifostine preserves osteocyte number and osteoid formation in fracture healing following radiotherapy. J. Oral. Maxillofac. Surg. 72, 559–566 (2014).

61

Harris, S. R. Differentiating the causes of spontaneous rib fracture after breast cancer. Clin. Breast Cancer 16, 431–436 (2016).

62

Kim, D., Kim, J. S., Shin, K. H. & Kim, K. Spontaneous rib fractures after breast cancer treatment based on bone scan: focusing on the radiotherapy. Int. J. Radiat. Oncol. 108, e30–e31 (2020).

63

Kim, D. W., Kim, J. S., Kim, K. & Shin, K. H. Spontaneous rib fractures after breast cancer treatment based on bone scans: comparison of conventional versus hypofractionated radiotherapy. Clin. Breast Cancer 21, e80–e87 (2021).

64

Oh, D. et al. Pelvic insufficiency fracture after pelvic radiotherapy for cervical cancer: analysis of risk factors. Int. J. Radiat. Oncol. 70, 1183–1188 (2008).

65

Paulino, A. C. Late effects of radiotherapy for pediatric extremity sarcomas. Int. J. Radiat. Oncol. 60, 265–274 (2004).

66

Zhai, J. et al. Influence of radiation exposure pattern on the bone injury and osteoclastogenesis in a rat model. Int. J. Mol. Med. 2019, 2265–2275 (2019).

67

Wright, L. E. et al. Single-limb irradiation induces local and systemic bone loss in a murine model. J. Bone Min. Res. 30, 1268–1279 (2015).

68

Hashem, R., Tanzer, M., Rene, N., Evans, M. & Souhami, L. Postoperative radiation therapy after hip replacement in high-risk patients for development of heterotopic bone formation. Cancer Radiothér. 15, 261–264 (2011).

69

Sibonga, J. D. Spaceflight-induced Bone Loss: Is there an Osteoporosis Risk? Curr. Osteoporos. Rep. 11, 92–98 (2013).

70

Orwoll, E. S. et al. Skeletal health in long-duration astronauts: Nature, assessment, and management recommendations from the NASA bone summit. J. Bone Min. Res. 28, 1243–1255 (2013).

71

Ishii, T. et al. A report from Fukushima: an assessment of bone health in an area affected by the Fukushima nuclear plant incident. J. Bone Min. Metab. 31, 613–617 (2013).

72

Masunari, N., Fujiwara, S., Nakata, Y., Nakashima, E. & Nakamura, T. Historical height loss, vertebral deformity, and health-related quality of life in Hiroshima cohort study. Osteoporos. Int. 18, 1493–1499 (2007).

73

Samartzis, D. et al. Exposure to ionizing radiation and development of bone sarcoma: new insights based on atomic-bomb survivors of Hiroshima and Nagasaki. J. Bone Joint Surg. Am. 93, 1008–1015 (2011).

74

Costa, S. & Reagan, M. R. Therapeutic irradiation: consequences for bone and bone marrow adipose tissue. Front. Endocrinol. (Lausanne). 10, 587 (2019).

75

Pieper, A. A. et al. Discovery of a proneurogenic, neuroprotective chemical. Cell 142, 39–51 (2010).

76

Borrego-Soto, G., Ortiz-López, R. & Rojas-Martínez, A. Ionizing radiation-induced DNA injury and damage detection in patients with breast cancer. Genet. Mol. Biol. 38, 420–432 (2015).

77

Tsuchida, E. et al. Effect of X-irradiation at different stages in the cell cycle on individual cell–based kinetics in an asynchronous cell population. PLoS One 10, e0128090 (2015).

78

Nelson, G., Kucheryavenko, O., Wordsworth, J. & von Zglinicki, T. The senescent bystander effect is caused by ROS-activated NF-κB signalling. Mech. Ageing Dev. 170, 30–36 (2018).

79

Bai, J. et al. Irradiation-induced senescence of bone marrow mesenchymal stem cells aggravates osteogenic differentiation dysfunction via paracrine signaling. Am. J. Physiol. Physiol. 318, C1005–C1017 (2020).

80

Cmielova, J. et al. Gamma radiation induces senescence in human adult mesenchymal stem cells from bone marrow and periodontal ligaments. Int. J. Radiat. Biol. 88, 393–404 (2012).

81

Meng, Q.-S. et al. Senescent mesenchymal stem/stromal cells and restoring their cellular functions. World J. Stem Cells 12, 966–985 (2020).

82

Panganiban, R.-A., Snow, A. & Day, R. Mechanisms of radiation toxicity in transformed and non-transformed cells. Int. J. Mol. Sci. 14, 15931–15958 (2013).

83

Escribano-Díaz, C. et al. A cell cycle-dependent regulatory circuit composed of 53BP1-RIF1 and BRCA1-CtIP controls DNA repair pathway choice. Mol. Cell 49, 872–883 (2013).

84

Levine, A. J. p53, the cellular gatekeeper for growth and division. Cell 88, 323–331 (1997).

85

Elbakrawy, E. M., Mayah, A., Hill, M. A. & Kadhim, M. Induction of genomic instability in a primary human fibroblast cell line following low-dose alpha-particle exposure and the potential role of exosomes. Biology 10, 11 (2020).

86

JAMALI, M. Persistent increase in the rates of apoptosis and dicentric chromosomes in surviving V79 cells after X-irradiation. Int. J. Radiat. Biol. 70, 705–709 (1996).

87

Chang, W. P. & Little, J. B. Persistently elevated frequency of spontaneous mutations in progeny of CHO clones surviving X-irradiation: association with delayed reproductive death phenotype. Mutat. Res. Mol. Mech. Mutagen. 270, 191–199 (1992).

88

Chang, W. P. & Little, J. B. Delayed reproductive death in X-irradiated Chinese hamster ovary cells. Int. J. Radiat. Biol. 60, 483–496 (1991).

89

Holmberg, K., Meijer, A. E., Auer, G. & Lambert, B. Delayed chromosomal instability in human T-lymphocyte clones exposed to ionizing radiation. Int. J. Radiat. Biol. 68, 245–255 (1995).

90

Moerman, E. J., Teng, K., Lipschitz, D. A. & Lecka-Czernik, B. Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR-γ2 transcription factor and TGF-β/BMP signaling pathways. Aging Cell 3, 379–389 (2004).

91

Muruganandan, S., Govindarajan, R. & Sinal, C. J. Bone marrow adipose tissue and skeletal health. Curr. Osteoporos. Rep. 16, 434–442 (2018).

92

McGee-Lawrence, M. E. et al. Hdac3 deficiency increases marrow adiposity and induces lipid storage and glucocorticoid metabolism in osteochondroprogenitor cells. J. Bone Min. Res. 31, 116–128 (2016).

93

Cohen, A. et al. Increased marrow adiposity in premenopausal women with idiopathic osteoporosis. J. Clin. Endocrinol. Metab. 97, 2782–2791 (2012).

94

Misra, M. & Klibanski, A. Anorexia nervosa, obesity and bone metabolism. Pediatr. Endocrinol. Rev. 11, 21–33 (2013).

95

Georgiou, K. R., Hui, S. K. & Xian, C. J. Regulatory pathways associated with bone loss and bone marrow adiposity caused by aging, chemotherapy, glucocorticoid therapy and radiotherapy. Am. J. Stem Cells 1, 205–224 (2012).

96

Chen, Q. et al. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ. 23, 1128–1139 (2016).

97

Li, J., Kwong, D. L. W. & Chan, G. C.-F. The effects of various irradiation doses on the growth and differentiation of marrow-derived human mesenchymal stromal cells. Pediatr. Transpl. 11, 379–387 (2007).

98

Sakurai, T., Sawada, Y., YOSHIMOTO, M., Kawai, M. & MIYAKOSHI, J. Radiation-induced reduction of osteoblast differentiation in C2C12 cells. J. Radiat. Res. 48, 515–521 (2007).

99

Bagchi, D. P. et al. Wnt/β-catenin signaling regulates adipose tissue lipogenesis and adipocyte-specific loss is rigorously defended by neighboring stromal-vascular cells. Mol. Metab. 42, 101078 (2020).

100

Chang, M.-K. et al. Disruption of Lrp4 function by genetic deletion or pharmacological blockade increases bone mass and serum sclerostin levels. Proc. Natl. Acad. Sci. 111, E5187–E5195 (2014).

101

Baroi, S., Czernik, P. J., Chougule, A., Griffin, P. R. & Lecka-Czernik, B. PPARG in osteocytes controls sclerostin expression, bone mass, marrow adiposity and mediates TZD-induced bone loss. Bone 147, 115913 (2021).

102

Kim, S. P. et al. Sclerostin influences body composition by regulating catabolic and anabolic metabolism in adipocytes. Proc. Natl. Acad. Sci. 114, E11238–E11247 (2017).

103

Ukita, M., Yamaguchi, T., Ohata, N. & Tamura, M. Sclerostin enhances adipocyte differentiation in 3T3‐L1 cells. J. Cell Biochem. 117, 1419–1428 (2016).

104

Bullock, W. A. et al. Lrp4 mediates bone homeostasis and mechanotransduction through interaction with sclerostin in vivo. iScience 20, 205–215 (2019).

105

Sakai, D. et al. Remodeling of actin cytoskeleton in mouse periosteal cells under mechanical loading induces periosteal cell proliferation during bone formation. PLoS One 6, e24847 (2011).

106

Chen, L. et al. Inhibiting actin depolymerization enhances osteoblast differentiation and bone formation in human stromal stem cells. Stem Cell Res. 15, 281–289 (2015).

107

Sen, B. et al. Intranuclear actin regulates osteogenesis. Stem Cells 33, 3065–3076 (2015).

108

Tong, J. et al. Cell micropatterning reveals the modulatory effect of cell shape on proliferation through intracellular calcium transients. Biochim Biophys. Acta Mol. Cell Res. 1864, 2389–2401 (2017).

109

Li, R. et al. Mechanical strain regulates osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells. Biomed. Res. Int. 2015, 1–10 (2015).

110

Kilian, K. A., Bugarija, B., Lahn, B. T. & Mrksich, M. Geometric cues for directing the differentiation of mesenchymal stem cells. Proc. Natl. Acad. Sci. 107, 4872–4877 (2010).

111

Górska, A. & Mazur, A. J. Integrin-linked kinase (ILK): the known vs. the unknown and perspectives. Cell Mol. Life Sci. 79, 100 (2022).

112

Andersson, O., Korach-Andre, M., Reissmann, E., Ibáñez, C. F. & Bertolino, P. Growth/differentiation factor 3 signals through ALK7 and regulates accumulation of adipose tissue and diet-induced obesity. Proc. Natl. Acad. Sci. 105, 7252–7256 (2008).

113

Hu, X. et al. Brd4 modulates diet-induced obesity via PPARγ-dependent Gdf3 expression in adipose tissue macrophages. JCI Insight. 6, e143379 (2021).

114

Witthuhn, B. A. & Bernlohr, D. A. Upregulation of bone morphogenetic protein GDF-3/Vgr-2 expression in adipose tissue of FABP4/aP2 null mice. Cytokine 14, 129–135 (2001).

115

Yahyapour, R. et al. Radiation-induced inflammation and autoimmune diseases. Mil. Med. Res. 5, 9 (2018).

116

Amarasekara, D. S., et al. Regulation of osteoclast differentiation by cytokine networks. Immune Netw. 18, e8 (2018).

117

Kowalski, E. J. A., Li, L. Toll-interacting protein in resolving and non-resolving inflammation. Front. Immunol. 8, 511 (2017).

118

Li, X., Goobie, G. C. & Zhang, Y. Toll-interacting protein impacts on inflammation, autophagy, and vacuole trafficking in human disease. J. Mol. Med. 99, 21–31 (2021).

119

Qayyum, N., Haseeb, M., Kim, M. S. & Choi, S. Role of thioredoxin-interacting protein in diseases and its therapeutic outlook. Int. J. Mol. Sci. 22, 2754 (2021).

120

Pi, C. et al. Nicotinamide phosphoribosyltransferase postpones rat bone marrow mesenchymal stem cell senescence by mediating NAD+–Sirt1 signaling. Aging (Albany NY) 11, 3505–3522 (2019).

121

He, X. et al. Nicotinamide phosphoribosyltransferase (Nampt) may serve as the marker for osteoblast differentiation of bone marrow-derived mesenchymal stem cells. Exp. Cell Res. 352, 45–52 (2017).

122

Romanello, M. et al. Extracellular NAD+: a novel autocrine/paracrine signal in osteoblast physiology. Biochem. Biophys. Res. Commun. 299, 424–431 (2002).

123

Iqbal, J. & Zaidi, M. Extracellular NAD+ metabolism modulates osteoclastogenesis. Biochem. Biophys. Res. Commun. 349, 533–539 (2006).

124

Li, B. et al. Attenuates of NAD+ impair BMSC osteogenesis and fracture repair through OXPHOS. Stem Cell Res. Ther. 13, 77 (2022).

125

Chen, W. et al. The neurogenic compound P7C3 regulates the aerobic glycolysis by targeting phosphoglycerate kinase 1 in Glioma. Front. Oncol. 11, 644492 (2021).

126

Ooi, A. T. & Gomperts, B. N. Molecular pathways: targeting cellular energy metabolism in cancer via inhibition of SLC2A1 and LDHA. Clin. Cancer Res. 21, 2440–2444 (2015).

127

Papaldo, P. et al. Addition of either lonidamine or granulocyte colony-stimulating factor does not improve survival in early breast cancer patients treated with high-dose epirubicin and cyclophosphamide. J. Clin. Oncol. 21, 3462–3468 (2003).

128

Vander Heiden, M. G. et al. Identification of small molecule inhibitors of pyruvate kinase M2. Biochem. Pharm. 79, 1118–1124 (2010).

129

Rani, R. & Kumar, V. Recent update on human lactate dehydrogenase enzyme 5 (h LDH5) inhibitors: a promising approach for cancer chemotherapy. J. Med. Chem. 59, 487–496 (2016).

130

Davies, J. H., Evans, B. A. J., Jenney, M. E. M. & Gregory, J. W. Effects of chemotherapeutic agents on the function of primary human osteoblast-like cells derived from children. J. Clin. Endocrinol. Metab. 88, 6088–6097 (2003).

131

Donaubauer, A.-J, et al. The influence of radiation on bone and bone cells-differential effects on osteoclasts and osteoblasts. Int. J. Mol. Sci. 21, 6377 (2020).

132

Belley, M. D. et al. Toward an organ based dose prescription method for the improved accuracy of murine dose in orthovoltage x-ray irradiators. Med. Phys. 41, 034101 (2014).

133

Wei, F., et al Multi-functional cerium oxide nanoparticles regulate inflammation and enhance osteogenesis. Mater Sci. Eng. C Mater. Biol. Appl. 124, 112041 (2021).

134

Jamsa, T., Jalovaara, P., Peng, Z., Vaananen, H. & Tuukkanen, J. Comparison of three-point bending test and peripheral quantitative computed tomography analysis in the evaluation of the strength of mouse femur and tibia. Bone 23, 155–161 (1998).

135

Schriefer, J. L. et al. A comparison of mechanical properties derived from multiple skeletal sites in mice. J. Biomech. 38, 467–475 (2005).

136

Omer, M. et al. Omega-9 modifies viscoelasticity and augments bone strength and architecture in a high-fat diet-fed murine model. Nutrients 14, 3165 (2022).

137

Deckard, C., Walker, A. & Hill, B. J. F. Using three-point bending to evaluate tibia bone strength in ovariectomized young mice. J. Biol. Phys. 43, 139–148 (2017).

138

Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

Bone Research
Article number: 34
Cite this article:
Wei F, Tuong ZK, Omer M, et al. A novel multifunctional radioprotective strategy using P7C3 as a countermeasure against ionizing radiation-induced bone loss. Bone Research, 2023, 11: 34. https://doi.org/10.1038/s41413-023-00273-w

146

Views

1

Downloads

5

Crossref

5

Web of Science

5

Scopus

Altmetrics

Received: 03 January 2023
Revised: 16 May 2023
Accepted: 28 May 2023
Published: 29 June 2023
© The Author(s) 2023

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

Return