Cell communication and relevant signaling pathways in osteogenesis–angiogenesis coupling

Shuqing Li , Xinjia Cai , Jiahe Guo , Xiaolu Li , Wen Li , Yan Liu , Mengchun Qi

Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 45

PDF
Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 45 DOI: 10.1038/s41413-025-00417-0
Review Article

Cell communication and relevant signaling pathways in osteogenesis–angiogenesis coupling

Author information +
History +
PDF

Abstract

Osteogenesis is the process of bone formation mediated by the osteoblasts, participating in various bone-related physiological processes including bone development, bone homeostasis and fracture healing. It exhibits temporal and spatial interconnectivity with angiogenesis, constructed by multiple forms of cell communication occurring between bone and vascular endothelial cells. Molecular regulation among different cell types is crucial for coordinating osteogenesis and angiogenesis to facilitate bone remodeling, fracture healing, and other bone-related processes. The transmission of signaling molecules and the activation of their corresponding signal pathways are indispensable for various forms of cell communication. This communication acts as a “bridge” in coupling osteogenesis to angiogenesis. This article reviews the modes and processes of cell communication in osteogenesis-angiogenesis coupling over the past decade, mainly focusing on interactions among bone-related cells and vascular endothelial cells to provide insights into the mechanism of cell communication of osteogenesis-angiogenesis coupling in different bone-related contexts. Moreover, clinical relevance and applications are also introduced in this review.

Keywords

Biological Sciences / Biochemistry and Cell Biology

Cite this article

Download citation ▾
Shuqing Li, Xinjia Cai, Jiahe Guo, Xiaolu Li, Wen Li, Yan Liu, Mengchun Qi. Cell communication and relevant signaling pathways in osteogenesis–angiogenesis coupling. Bone Research, 2025, 13(1): 45 DOI:10.1038/s41413-025-00417-0

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

StegenS, van GastelN, CarmelietG. Bringing new life to damaged bone: the importance of angiogenesis in bone repair and regeneration. Bone, 2015, 70: 19-27.

[2]

GrellierM, BordenaveL, AmedeeJ. Cell-to-cell communication between osteogenic and endothelial lineages: implications for tissue engineering. Trends Biotechnol., 2009, 27: 562-571.

[3]

ChenM, et al. . Skeleton-vasculature chain reaction: a novel insight into the mystery of homeostasis. Bone Res., 2021, 9: 21.

[4]

ArmingolE, OfficerA, HarismendyO, LewisNE. Deciphering cell-cell interactions and communication from gene expression. Nat. Rev. Genet., 2021, 22: 71-88.

[5]

JinL, LongY, ZhangQ, LongJ. MiRNAs regulate cell communication in osteogenesis-angiogenesis coupling during bone regeneration. Mol. Biol. Rep., 2023, 50: 8715-8728.

[6]

SuJ, et al. . Cell-cell communication: new insights and clinical implications. Signal Transduct. Target Ther., 2024, 9: 196.

[7]

JoinerDM, LessKD, Van WierenEM, HessD, WilliamsBO. Heterozygosity for an inactivating mutation in low-density lipoprotein-related receptor 6 (Lrp6) increases osteoarthritis severity in mice after ligament and meniscus injury. Osteoarthr. Cartil., 2013, 21: 1576-1585.

[8]

DeligiorgiMV, PanayiotidisMI, SiasosG, TrafalisDT. Osteoporosis entwined with cardiovascular disease: the implication of osteoprotegerin and the example of statins. Curr. Med. Chem., 2021, 28: 1443-1467.

[9]

ZhangD, et al. . Synaptic-like transmission between neural axons and arteriolar smooth muscle cells drives cerebral neurovascular coupling. Nat. Neurosci., 2024, 27: 232-248.

[10]

QinQ, et al. . Neurovascular coupling in bone regeneration. Exp. Mol. Med., 2022, 54: 1844-1849.

[11]

LiuW, et al. . Magnesium promotes bone formation and angiogenesis by enhancing MC3T3-E1 secretion of PDGF-BB. Biochem. Biophys. Res. Commun., 2020, 528: 664-670.

[12]

MattesB, ScholppS. Emerging role of contact-mediated cell communication in tissue development and diseases. Histochem. Cell Biol., 2018, 150: 431-442.

[13]

LiuQ, et al. . A proximity-tagging system to identify membrane protein–protein interactions. Nat. Methods, 2018, 15: 715-722.

[14]

VillarsF, et al. . Effect of HUVEC on human osteoprogenitor cell differentiation needs heterotypic gap junction communication. Am. J. Physiol. Cell Physiol., 2002, 282: C775-C785.

[15]

HautefortA, PfennigerA, KwakBR. Endothelial connexins in vascular function. Vasc. Biol., 2019, 1: H117-h124.

[16]

KoeppleC, et al. . Expression of Connexin43 stimulates endothelial angiogenesis independently of gap junctional communication in vitro. Int. J. Mol. Sci., 2021, 22: 7400.

[17]

MannellH, et al. . Cx43 promotes endothelial cell migration and angiogenesis via the tyrosine phosphatase SHP-2. Int. J. Mol. Sci., 2021, 23: 294.

[18]

FujitaK, XingQ, KhoslaS, MonroeDG. Mutual enhancement of differentiation of osteoblasts and osteocytes occurs through direct cell-cell contact. J. Cell Biochem., 2014, 115: 2039-2044

[19]

IshikawaM, et al. . Pannexin 3 ER Ca2+ channel gating is regulated by phosphorylation at the Serine 68 residue in osteoblast differentiation. Sci. Rep., 2019, 9. 18759

[20]

AstigianoC, et al. . Sirtuin 6 regulates the activation of the ATP/purinergic axis in endothelial cells. Int. J. Mol. Sci., 2023, 24: 6759.

[21]

LuoZ, et al. . Notch signaling in osteogenesis, osteoclastogenesis, and angiogenesis. Am. J. Pathol., 2019, 189: 1495-1500.

[22]

WangF, et al. . Mesenchymal cell-derived juxtacrine Wnt1 signaling regulates osteoblast activity and osteoclast differentiation. J. Bone Miner. Res., 2019, 34: 1129-1142.

[23]

GarciaMA, NelsonWJ, ChavezN. Cell-cell junctions organize structural and signaling networks. Cold Spring Harb. Perspect. Biol., 2018, 10: a029181.

[24]

WautierJL, WautierMP. Vascular permeability in diseases. Int. J. Mol. Sci., 2022, 23: 3645.

[25]

LampugnaniMG, OrsenigoF, GaglianiMC, TacchettiC, DejanaE. Vascular endothelial cadherin controls VEGFR-2 internalization and signaling from intracellular compartments. J. Cell Biol., 2006, 174: 593-604.

[26]

WangH, et al. . The osteogenic niche promotes early-stage bone colonization of disseminated breast cancer cells. Cancer Cell, 2015, 27: 193-210.

[27]

WatsonEC, AdamsRH. Biology of bone: the vasculature of the skeletal system. Cold Spring Harbor Perspect. Med., 2018, 8: a031559.

[28]

DuanX, et al. . Vegfa regulates perichondrial vascularity and osteoblast differentiation in bone development. Development, 2015, 142: 1984-1991.

[29]

HuK, OlsenBR. Osteoblast-derived VEGF regulates osteoblast differentiation and bone formation during bone repair. J. Clin. Invest., 2016, 126: 509-526.

[30]

WangW, et al. . Circ_0008542 in osteoblast exosomes promotes osteoclast-induced bone resorption through m6A methylation. Cell Death Dis., 2021, 12: 628.

[31]

WuC, et al. . Exosome miR-23a-3p from osteoblast alleviates spinal cord ischemia/reperfusion injury by down-regulating KLF3-activated CCNL2 transcription. Dev. Neurosci., 2022, 44: 121-130.

[32]

YangJX, XieP, LiYS, WenT, YangXC. Osteoclast-derived miR-23a-5p-containing exosomes inhibit osteogenic differentiation by regulating Runx2. Cell Signal, 2020, 70: 109504.

[33]

YueKY, et al. . Neurons can upregulate Cav-1 to increase intake of endothelial cells-derived extracellular vesicles that attenuate apoptosis via miR-1290. Cell Death Dis., 2019, 10: 869.

[34]

PanB, et al. . Macrophage-derived exosomes modulate wear particle-induced osteolysis via miR-3470b targeting TAB3/NF-kappaB signaling. Bioact. Mater., 2023, 26: 181-193

[35]

ZhangC, et al. . Monocytes deposit migrasomes to promote embryonic angiogenesis. Nat. Cell Biol., 2022, 24: 1726-1738.

[36]

SongH, et al. . Reversal of osteoporotic activity by endothelial cell-secreted bone targeting and biocompatible exosomes. Nano Lett., 2019, 19: 3040-3048.

[37]

CuiY, et al. . EPC-derived exosomes promote osteoclastogenesis through LncRNA-MALAT1. J. Cell Mol. Med., 2019, 23: 3843-3854.

[38]

BeheraJ, KumarA, VoorMJ, TyagiN. Exosomal lncRNA-H19 promotes osteogenesis and angiogenesis through mediating Angpt1/Tie2-NO signaling in CBS-heterozygous mice. Theranostics, 2021, 11: 7715-7734.

[39]

ZhaoX, et al. . Identification of markers for migrasome detection. Cell Discov., 2019, 5: 27.

[40]

JiangY, et al. . Migrasomes, a new mode of intercellular communication. Cell Commun. Signal., 2023, 21: 105.

[41]

YamamotoN, et al. . Rac limits TGF-beta-induced VEGF synthesis in osteoblasts. Mol. Cell Endocrinol., 2015, 405: 35-41.

[42]

JaradM, KuczynskiEA, MorrisonJ, Viloria-PetitAM, CoomberBL. Release of endothelial cell-associated VEGFR2 during TGF-beta modulated angiogenesis in vitro. BMC Cell Biol., 2017, 18. 10

[43]

HiepenC, et al. . BMPR2 acts as a gatekeeper to protect endothelial cells from increased TGFβ responses and altered cell mechanics. PLoS Biol., 2019, 17: e3000557.

[44]

WangX, JiL, WangJ, LiuC. Matrix stiffness regulates osteoclast fate through integrin-dependent mechanotransduction. Bioact. Mater., 2023, 27: 138-153

[45]

TangY, et al. . Phosphorylation inhibition of protein-tyrosine phosphatase 1B tyrosine-152 induces bone regeneration coupled with angiogenesis for bone tissue engineering. Bioact. Mater., 2021, 6: 2039-2057

[46]

GenovaT, et al. . TRPM8 inhibits endothelial cell migration via a non-channel function by trapping the small GTPase Rap1. J. Cell Biol., 2017, 216: 2107-2130.

[47]

LindemannO, et al. . Intravascular adhesion and recruitment of neutrophils in response to CXCL1 depends on their TRPC6 channels. J. Mol. Med., 2020, 98: 349-360.

[48]

Garbuzova-DavisS, et al. . Cell-free extracellular vesicles derived from human bone marrow endothelial progenitor cells as potential therapeutics for microvascular endothelium restoration in ALS. Neuromol. Med., 2020, 22: 503-516.

[49]

DanilucciTM, et al. . Recombinant RGD-disintegrin DisBa-01 blocks integrin alpha(v)beta(3) and impairs VEGF signaling in endothelial cells. Cell Commun. Signal., 2019, 17: 27.

[50]

GiordanoA, et al. . Tirofiban counteracts endothelial cell apoptosis through the VEGF/VEGFR2/pAkt axis. Vasc. Pharm., 2016, 80: 67-74.

[51]

HusainA, KhadkaA, EhrlicherA, Saint-GeniezM, KrishnanR. Substrate stiffening promotes VEGF-A functions via the PI3K/Akt/mTOR pathway. Biochem. Biophys. Res. Commun., 2022, 586: 27-33.

[52]

KuriyamaM, et al. . Activation and translocation of PKCdelta is necessary for VEGF-induced ERK activation through KDR in HEK293T cells. Biochem. Biophys. Res. Commun., 2004, 325: 843-851.

[53]

PhoenixKN, et al. . PLCbeta2 promotes VEGF-induced vascular permeability. Arterioscler. Thromb. Vasc. Biol., 2022, 42: 1229-1241.

[54]

ShenJ, RossatoFA, CanoI, NgYSE. Novel engineered, membrane-tethered VEGF-A variants promote formation of filopodia, proliferation, survival, and cord or tube formation by endothelial cells via persistent VEGFR2/ERK signaling and activation of CDC42/ROCK pathways. FASEB J., 2021, 35: e22036.

[55]

LamaliceL, HouleF, JourdanG, HuotJ. Phosphorylation of tyrosine 1214 on VEGFR2 is required for VEGF-induced activation of Cdc42 upstream of SAPK2/p38. Oncogene, 2004, 23: 434-445.

[56]

BarryDM, et al. . Cdc42 is required for cytoskeletal support of endothelial cell adhesion during blood vessel formation in mice. Development, 2015, 142: 3058-3070

[57]

SjobergE, et al. . Endothelial VEGFR2-PLCgamma signaling regulates vascular permeability and antitumor immunity through eNOS/Src. J. Clin. Investig., 2023, 133: e161366.

[58]

GuoGX, et al. . The extract of Curcumae Longae Rhizoma suppresses angiogenesis via VEGF-induced PI3K/Akt-eNOS-NO pathway. J. Ethnopharmacol., 2023, 308: 116299.

[59]

LiH, et al. . FGA controls VEGFA secretion to promote angiogenesis by activating the VEGFR2-FAK signalling pathway. Front. Endocrinol., 2022, 13: 791860.

[60]

HayashiM, et al. . VE-PTP regulates VEGFR2 activity in stalk cells to establish endothelial cell polarity and lumen formation. Nat. Commun., 2013, 4. 1672

[61]

LeeC, et al. . VEGF-B prevents excessive angiogenesis by inhibiting FGF2/FGFR1 pathway. Signal Transduct. Target Ther., 2023, 8: 305.

[62]

StreetJ, et al. . Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc. Natl. Acad. Sci. USA, 2002, 99: 9656-9661.

[63]

da Silva SassoGR, et al. . Spatio-temporal immunolocalization of VEGF-A, Runx2, and osterix during the early steps of intramembranous ossification of the alveolar process in rat embryos. Dev. Biol., 2021, 478: 133-143.

[64]

BouletreauPJ, et al. . Hypoxia and VEGF up-regulate BMP-2 mRNA and protein expression in microvascular endothelial cells: implications for fracture healing. Plast. Reconstr. Surg., 2002, 109: 2384-2397.

[65]

MotokawaM, et al. . Effects of vascular endothelial growth factor-C and -D on osteoclast differentiation and function in human peripheral blood mononuclear cells. Arch. Oral. Biol., 2013, 58: 35-41.

[66]

CockmanME, et al. . Hypoxia inducible factor-alpha binding and ubiquitylation by the von Hippel-Lindau tumor suppressor protein. J. Biol. Chem., 2000, 275: 25733-25741.

[67]

KimHH, et al. . Stabilization of hypoxia-inducible factor-1alpha is involved in the hypoxic stimuli-induced expression of vascular endothelial growth factor in osteoblastic cells. Cytokine, 2002, 17: 14-27.

[68]

WangY, et al. . The hypoxia-inducible factor alpha pathway couples angiogenesis to osteogenesis during skeletal development. J. Clin. Investig., 2007, 117: 1616-1626.

[69]

WanC, et al. . Activation of the hypoxia-inducible factor-1alpha pathway accelerates bone regeneration. Proc. Natl. Acad. Sci. USA, 2008, 105: 686-691.

[70]

WangJ, et al. . CD31hiEmcnhi vessels support new trabecular bone formation at the frontier growth area in the bone defect repair process. Sci. Rep., 2017, 7. 4990

[71]

ChenG, DengC, LiYP. TGF-β and BMP signaling in osteoblast differentiation and bone formation. Int. J. Biol. Sci., 2012, 8: 272-288.

[72]

SampathTK, ReddiAH. Discovery of bone morphogenetic proteins—a historical perspective. Bone, 2020, 140: 115548.

[73]

SampathTK, VukicevicS. Biology of bone morphogenetic protein in bone repair and regeneration: a role for autologous blood coagulum as carrier. Bone, 2020, 141: 115602.

[74]

KimJM, LinC, StavreZ, GreenblattMB, ShimJH. Osteoblast-osteoclast communication and bone homeostasis. Cells, 2020, 9: 2073.

[75]

Alcorta-SevillanoN, MaciasI, InfanteA, RodriguezCI. Deciphering the relevance of bone ECM signaling. Cells, 2020, 9: 2630.

[76]

LuoK. Signaling cross-talk between TGF-β/Smad and other signaling pathways. Cold Spring Harb. Perspect. Biol., 2017, 9: a022137.

[77]

ZouML, et al. . The Smad dependent TGF-β and BMP signaling pathway in bone remodeling and therapies. Front. Mol. Biosci., 2021, 8: 593310.

[78]

RahmanMS, AkhtarN, JamilHM, BanikRS, AsaduzzamanSM. TGF-β/BMP signaling and other molecular events: regulation of osteoblastogenesis and bone formation. Bone Res., 2015, 3: 15005.

[79]

ZouML, et al. . The Smad-dependent TGF-beta and BMP signaling pathway in bone remodeling and therapies. Front. Mol. Biosci., 2021, 8: 593310.

[80]

CaoZ, LiuG, ZhangH, WangM, XuY. Nox4 promotes osteoblast differentiation through TGF-beta signal pathway. Free Radic. Biol. Med., 2022, 193: 595-609.

[81]

AsaiK, HisasueM, ShimokawaF, FunabaM, MurakamiM. TGF-beta negatively regulates Mitf-E expression and canine osteoclastogenesis. Biochem. Genet., 2018, 56: 542-552.

[82]

ZhangY, YangW, DevitA, van den BeuckenJ. Efficiency of coculture with angiogenic cells or physiological BMP-2 administration on improving osteogenic differentiation and bone formation of MSCs. J. Biomed. Mater. Res. Part A, 2019, 107: 643-653.

[83]

SalhotraA, ShahHN, LeviB, LongakerMT. Mechanisms of bone development and repair. Nat. Rev. Mol. Cell Biol., 2020, 21: 696-711.

[84]

WangZ, et al. . Experimental study of the synergistic effect and network regulation mechanisms of an applied combination of BMP-2, VEGF, and TGF-beta1 on osteogenic differentiation. J. Cell Biochem., 2020, 121: 2394-2405.

[85]

RimEY, CleversH, NusseR. The Wnt pathway: from signaling mechanisms to synthetic modulators. Annu. Rev. Biochem., 2022, 91: 571-598.

[86]

HuL, ChenW, QianA, LiYP. Wnt/beta-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res., 2024, 12: 39.

[87]

KobayashiY, UeharaS, UdagawaN, TakahashiN. Regulation of bone metabolism by Wnt signals. J. Biochem., 2016, 159: 387-392.

[88]

YeX, LiuX. Wnt16 signaling in bone homeostasis and osteoarthristis. Front. Endocrinol., 2022, 13: 1095711.

[89]

ZhuP, et al. . Panax notoginseng saponins promote endothelial progenitor cell angiogenesis via the Wnt/beta-catenin pathway. BMC Complement. Med. Ther., 2021, 21: 53.

[90]

ZhengS, HuG, ZhengJ, LiY, LiJ. Osthole accelerates osteoporotic fracture healing by inducing the osteogenesis-angiogenesis coupling of BMSCs via the Wnt/beta-catenin pathway. Phytother. Res., 2024, 38: 4022-4035.

[91]

ShenJ, et al. . EGFL6 regulates angiogenesis and osteogenesis in distraction osteogenesis via Wnt/beta-catenin signaling. Stem Cell Res. Ther., 2021, 12: 415.

[92]

YuB, et al. . Wnt4 signaling prevents skeletal aging and inflammation by inhibiting nuclear factor-kappaB. Nat. Med., 2014, 20: 1009-1017.

[93]

YangT, et al. . Wnt5a/Ror2 mediates temporomandibular joint subchondral bone remodeling. J. Dent. Res., 2015, 94: 803-812.

[94]

Moverare-SkrticS, et al. . Osteoblast-derived WNT16 represses osteoclastogenesis and prevents cortical bone fragility fractures. Nat. Med., 2014, 20: 1279-1288.

[95]

KobayashiY, et al. . Wnt16 regulates osteoclast differentiation in conjunction with Wnt5a. Biochem. Biophys. Res. Commun., 2015, 463: 1278-1283.

[96]

BennettCN, et al. . Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc. Natl. Acad. Sci. USA, 2005, 102: 3324-3329.

[97]

XieY, et al. . FGF/FGFR signaling in health and disease. Signal Transduct. Target Ther., 2020, 5: 181.

[98]

ItohN. Hormone-like (endocrine) Fgfs: their evolutionary history and roles in development, metabolism, and disease. Cell Tissue Res., 2010, 342: 1-11.

[99]

BehrB, LeuchtP, LongakerMT, QuartoN. Fgf-9 is required for angiogenesis and osteogenesis in long bone repair. Proc. Natl. Acad. Sci. USA, 2010, 107: 11853-11858.

[100]

BehrB, et al. . Fgf-18 is required for osteogenesis but not angiogenesis during long bone repair. Tissue Eng. Part A, 2011, 17: 2061-2069.

[101]

FinettiF, et al. . Prostaglandin E2 regulates angiogenesis via activation of fibroblast growth factor receptor-1. J. Biol. Chem., 2008, 283: 2139-2146.

[102]

FinettiF, DonniniS, GiachettiA, MorbidelliL, ZicheM. Prostaglandin E(2) primes the angiogenic switch via a synergic interaction with the fibroblast growth factor-2 pathway. Circ. Res., 2009, 105: 657-666.

[103]

LeeKW, et al. . FGF11 induced by hypoxia interacts with HIF-1alpha and enhances its stability. FEBS Lett., 2017, 591: 348-357.

[104]

YangJ, et al. . Hypoxia-induced fibroblast growth factor 11 stimulates capillary-like endothelial tube formation. Oncol. Rep., 2015, 34: 2745-2751.

[105]

KnowlesHJ. Hypoxia-induced fibroblast growth factor 11 stimulates osteoclast-mediated resorption of bone. Calcif. Tissue Int., 2017, 100: 382-391.

[106]

ZhangX, XuJ. A novel miR-466l-3p/FGF23 axis promotes osteogenic differentiation of human bone marrow mesenchymal stem cells. Bone, 2024, 185: 117123.

[107]

ShenZ, et al. . Total flavonoids of rhizoma drynariae enhances angiogenic-osteogenic coupling during distraction osteogenesis by promoting type H vessel formation through PDGF-BB/PDGFR-beta instead of HIF-1alpha/ VEGF axis. Front. Pharm., 2020, 11: 503524.

[108]

GaoSY, et al. . PDGF-BB exhibited therapeutic effects on rat model of bisphosphonate-related osteonecrosis of the jaw by enhancing angiogenesis and osteogenesis. Bone, 2021, 144: 115117.

[109]

WangF, et al. . PDGFR in PDGF-BB/PDGFR signaling pathway does orchestrates osteogenesis in a temporal manner. Research, 2023, 6: 0086.

[110]

KusumbeAP, RamasamySK, AdamsRH. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature, 2014, 507: 323-328.

[111]

LangenUH, et al. . Cell-matrix signals specify bone endothelial cells during developmental osteogenesis. Nat. Cell Biol., 2017, 19: 189-201.

[112]

PengY, WuS, LiY, CraneJL. Type H blood vessels in bone modeling and remodeling. Theranostics, 2020, 10: 426-436.

[113]

DzamukovaM, et al. . Mechanical forces couple bone matrix mineralization with inhibition of angiogenesis to limit adolescent bone growth. Nat. Commun., 2022, 13. 3059

[114]

ZhangJ, PanJ, JingW. Motivating role of type H vessels in bone regeneration. Cell Prolif., 2020, 53: e12874.

[115]

RamasamySK, KusumbeAP, WangL, AdamsRH. Endothelial Notch activity promotes angiogenesis and osteogenesis in bone. Nature, 2014, 507: 376-380.

[116]

XieH, et al. . PDGF-BB secreted by preosteoclasts induces angiogenesis during coupling with osteogenesis. Nat. Med., 2014, 20: 1270-1278.

[117]

HuangJ, et al. . Harmine enhances type H vessel formation and prevents bone loss in ovariectomized mice. Theranostics, 2018, 8: 2435-2446.

[118]

LiuJH, et al. . Akkermansia muciniphila promotes type H vessel formation and bone fracture healing by reducing gut permeability and inflammation. Dis. Models Mech., 2020, 13: dmm043620.

[119]

XuS, et al. . Dexamethasone interferes with osteoblasts formation during osteogenesis through altering IGF-1-mediated angiogenesis. J. Cell Physiol., 2019, 234: 15167-15181.

[120]

WangT, et al. . Osteoblast-specific loss of IGF1R signaling results in impaired endochondral bone formation during fracture healing. J. Bone Min. Res, 2015, 30: 1572-1584.

[121]

QiX, LiX. Mechanistic insights into the generation and transduction of Hedgehog signaling. Trends Biochem. Sci., 2020, 45: 397-410.

[122]

DohleE, et al. . Sonic hedgehog promotes angiogenesis and osteogenesis in a coculture system consisting of primary osteoblasts and outgrowth endothelial cells. Tissue Eng. Part A, 2010, 16: 1235-1237.

[123]

RivronNC, et al. . Sonic Hedgehog-activated engineered blood vessels enhance bone tissue formation. Proc. Natl. Acad. Sci. USA, 2012, 109: 4413-4418.

[124]

KazmersNH, McKenzieJA, ShenTS, LongF, SilvaMJ. Hedgehog signaling mediates woven bone formation and vascularization during stress fracture healing. Bone, 2015, 81: 524-532.

[125]

LorthongpanichC, et al. . YAP as a key regulator of adipo-osteogenic differentiation in human MSCs. Stem Cell Res. Ther., 2019, 10: 402.

[126]

ZhouA, et al. . Role of hippo-YAP signaling in osseointegration by regulating osteogenesis, angiogenesis, and osteoimmunology. Front. Cell Dev. Biol., 2020, 8: 780.

[127]

WangB, et al. . Neural peptide promotes the angiogenesis and osteogenesis around oral implants. Cell Signal, 2021, 79: 109873.

[128]

GaoL, et al. . Targeting soluble epoxide hydrolase promotes osteogenic-angiogenic coupling via activating SLIT3/HIF-1alpha signalling pathway. Cell Prolif., 2023, 56: e13403.

[129]

XuR, et al. . Targeting skeletal endothelium to ameliorate bone loss. Nat. Med., 2018, 24: 823-833.

[130]

RamasamySK, et al. . Regulation of hematopoiesis and osteogenesis by blood vessel-derived signals. Annu. Rev. Cell Dev. Biol., 2016, 32: 649-675.

[131]

ArthurA, GronthosS. Eph-ephrin signaling mediates cross-talk within the bone microenvironment. Front. Cell Dev. Biol., 2021, 9: 598612.

[132]

ZhaoC, et al. . Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab., 2006, 4: 111-121.

[133]

ZhangY, WangXC, BaoXF, HuM, YuWX. Effects of porphyromonas gingivalis lipopolysaccharide on osteoblast-osteoclast bidirectional EphB4-EphrinB2 signaling. Exp. Ther. Med., 2014, 7: 80-84.

[134]

Hassan-MohamedI, et al. . UniPR129 is a competitive small molecule Eph-ephrin antagonist blocking in vitro angiogenesis at low micromolar concentrations. Br. J. Pharm., 2014, 171: 5195-5208.

[135]

MaesC, et al. . Impaired angiogenesis and endochondral bone formation in mice lacking the vascular endothelial growth factor isoforms VEGF164 and VEGF188. Mech. Dev., 2002, 111: 61-73.

[136]

MaesC, et al. . Increased skeletal VEGF enhances beta-catenin activity and results in excessively ossified bones. EMBO J., 2010, 29: 424-441.

[137]

MaesC, et al. . Soluble VEGF isoforms are essential for establishing epiphyseal vascularization and regulating chondrocyte development and survival. J. Clin. Investig., 2004, 113: 188-199.

[138]

BixelMG, et al. . Flow dynamics and HSPC homing in bone marrow microvessels. Cell Rep., 2017, 18: 1804-1816.

[139]

ItkinT, et al. . Distinct bone marrow blood vessels differentially regulate haematopoiesis. Nature, 2016, 532: 323-328.

[140]

ZhangJ, et al. . In situ mapping identifies distinct vascular niches for myelopoiesis. Nature, 2021, 590: 457-462.

[141]

ZhaoY, et al. . Crosstalk between the neuroendocrine system and bone homeostasis. Endocr. Rev., 2024, 45: 95-124.

[142]

RibattiD, d’AmatiA. Bone angiocrine factors. Front. Cell Dev. Biol., 2023, 11: 1244372.

[143]

JiangL, SunJ, HuangD. Role of Slit/Robo signaling pathway in bone metabolism. Int. J. Biol. Sci., 2022, 18: 1303-1312.

[144]

ZhaoY, XieL. Unique bone marrow blood vessels couple angiogenesis and osteogenesis in bone homeostasis and diseases. Ann. N.Y. Acad. Sci., 2020, 1474: 5-14.

[145]

ChenY, et al. . Occlusal force maintains alveolar bone homeostasis via Type H angiogenesis. J. Dent. Res., 2023, 102: 1356-1365.

[146]

MurphyJF, FitzgeraldDJ. Vascular endothelial growth factor induces cyclooxygenase-dependent proliferation of endothelial cells via the VEGF-2 receptor. FASEB J., 2001, 15: 1667-1669.

[147]

ClarkinCE, EmeryRJ, PitsillidesAA, Wheeler-JonesCP. Evaluation of VEGF-mediated signaling in primary human cells reveals a paracrine action for VEGF in osteoblast-mediated crosstalk to endothelial cells. J. Cell Physiol., 2008, 214: 537-544.

[148]

ShaoJ, et al. . A dual role of HIF1alpha in regulating osteogenesis-angiogenesis coupling. Stem Cell Res. Ther., 2022, 13: 59.

[149]

ZhaoSJ, et al. . Macrophage MSR1 promotes BMSC osteogenic differentiation and M2-like polarization by activating PI3K/AKT/GSK3beta/beta-catenin pathway. Theranostics, 2020, 10: 17-35.

[150]

ZhangY, ZhouL, FuQ, LiuZ. ANKRD1 activates the Wnt signaling pathway by modulating CAV3 expression and thus promotes BMSC osteogenic differentiation and bone formation in ovariectomized mice. Biochim. Biophys. Acta Mol. Basis Dis., 2023, 1869: 166693.

[151]

GengW, et al. . Substance P enhances BMSC osteogenic differentiation via autophagic activation. Mol. Med. Rep., 2019, 20: 664-670

[152]

LiJ, et al. . Type H vessel/platelet-derived growth factor receptor beta(+) perivascular cell disintegration is involved in vascular injury and bone loss in radiation-induced bone damage. Cell Prolif., 2023, 56: e13406.

[153]

ZhuS, et al. . Endothelial cells produce angiocrine factors to regulate bone and cartilage via versatile mechanisms. Theranostics, 2020, 10: 5957-5965.

[154]

StavriGT, et al. . Hypoxia and platelet-derived growth factor-BB synergistically upregulate the expression of vascular endothelial growth factor in vascular smooth muscle cells. FEBS Lett., 1995, 358: 311-315.

[155]

XuJ, et al. . Bone-forming perivascular cells: cellular heterogeneity and use for tissue repair. Stem Cells, 2021, 39: 1427-1434.

[156]

HuangB, et al. . Osteoblasts secrete Cxcl9 to regulate angiogenesis in bone. Nat. Commun., 2016, 7. 13885

[157]

SaxtonRA, SabatiniDM. mTOR signaling in growth, metabolism, and disease. Cell, 2017, 169: 361-371.

[158]

HuangB, et al. . mTORC1 prevents preosteoblast differentiation through the notch signaling pathway. PLoS Genet., 2015, 11: e1005426.

[159]

Tsuji-TamuraK, OgawaM. Inhibition of the PI3K-Akt and mTORC1 signaling pathways promotes the elongation of vascular endothelial cells. J. Cell Sci., 2016, 129: 1165-1178.

[160]

OberkerschRE, et al. . Aspartate metabolism in endothelial cells activates the mTORC1 pathway to initiate translation during angiogenesis. Dev. Cell, 2022, 57: 1241-1256 e1248.

[161]

ZieglerME, HatchMM, WuN, MuawadSA, HughesCC. mTORC2 mediates CXCL12-induced angiogenesis. Angiogenesis, 2016, 19: 359-371.

[162]

FarhanMA, Carmine-SimmenK, LewisJD, MooreRB, MurrayAG. Endothelial cell mTOR complex-2 regulates sprouting angiogenesis. PloS One, 2015, 10: e0135245.

[163]

ZhuN, et al. . Endothelin-1 triggers human peritoneal mesothelial cells’ proliferation via ERK1/2-Ets-1 signaling pathway and contributes to endothelial cell angiogenesis. J. Cell Biochem., 2019, 120: 3539-3546.

[164]

ZhongX, WangH, HuangS. Endothelin-1 induces interleukin-18 expression in human osteoblasts. Arch. Oral. Biol., 2014, 59: 289-296.

[165]

WangDS, MiuraM, DemuraH, SatoK. Anabolic effects of 1,25-dihydroxyvitamin D3 on osteoblasts are enhanced by vascular endothelial growth factor produced by osteoblasts and by growth factors produced by endothelial cells. Endocrinology, 1997, 138: 2953-2962.

[166]

DicarloM, et al. . Evidence supporting a paracrine effect of IGF-1/VEGF on human mesenchymal stromal cell commitment. Cells Tissues Organs, 2016, 201: 333-341.

[167]

HeY, et al. . Exosomal let-7f-5p derived from mineralized osteoblasts promotes the angiogenesis of endothelial cells via the DUSP1/Erk1/2 signaling pathway. J. Tissue Eng. Regener. Med., 2022, 16: 1184-1195.

[168]

GuoZ, et al. . Exosomal miR-214-3p from senescent osteoblasts accelerates endothelial cell senescence. J. Orthop. Surg. Res., 2023, 18: 391.

[169]

ZhangX, et al. . Exosomes from microvascular endothelial cells under mechanical unloading inhibit osteogenic differentiation via miR-92b-3p/ELK4 Axis. J. Pers. Med., 2022, 12: 2030.

[170]

YangRZ, et al. . Exosomes derived from vascular endothelial cells antagonize glucocorticoid-induced osteoporosis by inhibiting ferritinophagy with resultant limited ferroptosis of osteoblasts. J. Cell Physiol., 2021, 236: 6691-6705.

[171]

TangH, et al. . Exosomal MMP2 derived from mature osteoblasts promotes angiogenesis of endothelial cells via VEGF/Erk1/2 signaling pathway. Exp. Cell Res., 2019, 383: 111541.

[172]

WuH, et al. . Mechanism of vascular endothelial cell-derived exosomes modified with vascular endothelial growth factor in steroid-induced femoral head necrosis. Biomed. Mater., 2023, 18: 025017.

[173]

MiB, et al. . Osteoblast/osteoclast and immune cocktail therapy of an exosome/drug delivery multifunctional hydrogel accelerates fracture repair. ACS Nano, 2022, 16: 771-782.

[174]

WangQ, LuQ. Plasma membrane-derived extracellular microvesicles mediate non-canonical intercellular NOTCH signaling. Nat. Commun., 2017, 8. 709

[175]

Sharghi-NaminiS, TanE, OngLL, GeR, AsadaHH. Dll4-containing exosomes induce capillary sprout retraction in a 3D microenvironment. Sci. Rep., 2014, 4. 4031

[176]

GrellierM, et al. . Role of vascular endothelial growth factor in the communication between human osteoprogenitors and endothelial cells. J. Cell Biochem., 2009, 106: 390-398.

[177]

FanX, TengY, YeZ, ZhouY, TanWS. The effect of gap junction-mediated transfer of miR-200b on osteogenesis and angiogenesis in a co-culture of MSCs and HUVECs. J. Cell Sci., 2018, 131: jcs216135.

[178]

QinH, et al. . Magnesium ions promote in vitro rat bone marrow stromal cell angiogenesis through notch signaling. Biol. Trace Elem. Res., 2023, 201: 2823-2842.

[179]

LiL, et al. . GIT1 regulates angiogenic factor secretion in bone marrow mesenchymal stem cells via NF-kappaB/Notch signalling to promote angiogenesis. Cell Prolif., 2019, 52: e12689.

[180]

YangM, et al. . MiR-497 approximately 195 cluster regulates angiogenesis during coupling with osteogenesis by maintaining endothelial Notch and HIF-1alpha activity. Nat. Commun., 2017, 8. 16003

[181]

ClarkinCE, GerstenfeldLC. VEGF and bone cell signalling: an essential vessel for communication. Cell Biochem. Funct., 2013, 31: 1-11.

[182]

PitulescuME, et al. . Dll4 and Notch signalling couples sprouting angiogenesis and artery formation. Nat. Cell Biol., 2017, 19: 915-927.

[183]

HellstromM, et al. . Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature, 2007, 445: 776-780.

[184]

DandajenaTC, IhnatMA, DischB, ThorpeJ, CurrierGF. Hypoxia triggers a HIF-mediated differentiation of peripheral blood mononuclear cells into osteoclasts. Orthod. Craniofac. Res., 2012, 15: 1-9.

[185]

HulleyPA, et al. . Hypoxia-inducible factor 1-alpha does not regulate osteoclastogenesis but enhances bone resorption activity via prolyl-4-hydroxylase 2. J. Pathol., 2017, 242: 322-333.

[186]

KnowlesHJ. Distinct roles for the hypoxia-inducible transcription factors HIF-1alpha and HIF-2alpha in human osteoclast formation and function. Sci. Rep., 2020, 10. 21072

[187]

TaylorRM, KashimaTG, KnowlesHJ, AthanasouNA. VEGF, FLT3 ligand, PlGF and HGF can substitute for M-CSF to induce human osteoclast formation: implications for giant cell tumour pathobiology. Lab. Investig., 2012, 92: 1398-1406.

[188]

Trebec-ReynoldsDP, VoronovI, HeerscheJN, ManolsonMF. VEGF-A expression in osteoclasts is regulated by NF-kappaB induction of HIF-1alpha. J. Cell Biochem., 2010, 110: 343-351.

[189]

GroppaE, et al. . VEGF dose regulates vascular stabilization through Semaphorin3A and the Neuropilin-1+ monocyte/TGF-beta1 paracrine axis. EMBO Mol. Med., 2015, 7: 1366-1384.

[190]

LiuYQ, HanXF, BoJX, MaHP. Wedelolactone enhances osteoblastogenesis but inhibits osteoclastogenesis through Sema3A/NRP1/PlexinA1 pathway. Front. Pharm., 2016, 7: 375.

[191]

GuoZ, et al. . Semaphorin3A regulates mitochondrial apoptosis in RAW264.7 cells in vitro. Tissue Cell, 2022, 75: 101711.

[192]

GaoSY, et al. . Zoledronate suppressed angiogenesis and osteogenesis by inhibiting osteoclasts formation and secretion of PDGF-BB. PloS One, 2017, 12: e0179248.

[193]

XuT, et al. . GIT1 is critical for formation of the CD31(hi)Emcn(hi) vessel subtype in coupling osteogenesis with angiogenesis via modulating preosteoclasts secretion of PDGF-BB. Bone, 2019, 122: 218-230.

[194]

WangY, ZhengY, LiW. Compression loading of osteoclasts attenuated microRNA-146a-5p expression, which promotes angiogenesis by targeting adiponectin. Sci. China Life Sci., 2022, 65: 151-166.

[195]

CaiM, et al. . Vascular pericyte-derived exosomes inhibit bone resorption via Traf3. Int. J. Nanomed., 2023, 18: 7065-7077.

[196]

WiedemannE, et al. . Regulation of endothelial migration and proliferation by ephrin-A1. Cell Signal, 2017, 29: 84-95.

[197]

WangY, et al. . Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature, 2010, 465: 483-486.

[198]

PoitzDM, et al. . EphrinB2/EphA4-mediated activation of endothelial cells increases monocyte adhesion. Mol. Immunol., 2015, 68: 648-656.

[199]

SuzakiA, Komine-AizawaS, HayakawaS. Suppression of osteoblast toll-like receptor 2 signaling by endothelin-1. J. Orthop. Res., 2014, 32: 910-914.

[200]

SenS, LuxCJ, ErberR. A potential role of semaphorin 3A during orthodontic tooth movement. Int. J. Mol. Sci., 2021, 22: 8297.

[201]

UdagawaN, et al. . Osteoclast differentiation by RANKL and OPG signaling pathways. J. Bone Min. Metab., 2021, 39: 19-26.

[202]

DengL, et al. . Osteoblast-derived microvesicles: a novel mechanism for communication between osteoblasts and osteoclasts. Bone, 2015, 79: 37-42.

[203]

ChenC, ZhengRQ, CaoXC, ZhangGC. Biological characteristics of osteoclast exosomes and their role in the osteogenic differentiation of somatic cells prior to osteogenesis. J. Biol. Regul. Homeost. Agents, 2018, 32: 815-823

[204]

IkebuchiY, et al. . Coupling of bone resorption and formation by RANKL reverse signalling. Nature, 2018, 561: 195-200.

[205]

CraneJL, CaoX. Function of matrix IGF-1 in coupling bone resorption and formation. J. Mol. Med., 2014, 92: 107-115.

[206]

YakarS, WernerH, RosenCJ. Insulin-like growth factors: actions on the skeleton. J. Mol. Endocrinol., 2018, 61: T115-T137.

[207]

PaganiS, et al. . An advanced tri-culture model to evaluate the dynamic interplay among osteoblasts, osteoclasts, and endothelial cells. J. Cell Physiol., 2018, 233: 291-301.

[208]

LuoY, et al. . Immunomodulatory nanomedicine for osteoporosis: current practices and emerging prospects. Acta Biomater., 2024, 179: 13-35.

[209]

SunY, et al. . Macrophage-osteoclast associations: origin, polarization, and subgroups. Front. Immunol., 2021, 12: 778078.

[210]

HorwoodNJ. Macrophage polarization and bone formation: a review. Clin. Rev. Allergy Immunol., 2016, 51: 79-86.

[211]

HernandezGE, Iruela-ArispeML. The many flavors of monocyte/macrophage–endothelial cell interactions. Curr. Opin. Hematol., 2020, 27: 181-189.

[212]

DengZ, et al. . Crosstalk between immune cells and bone cells or chondrocytes. Int. Immunopharmacol., 2021, 101: 108179.

[213]

Khatib-MassalhaE, et al. . Lactate released by inflammatory bone marrow neutrophils induces their mobilization via endothelial GPR81 signaling. Nat. Commun., 2020, 11. 3547

[214]

AndoY, et al. . The neutrophil-osteogenic cell axis promotes bone destruction in periodontitis. Int. J. Oral. Sci., 2024, 16: 18.

[215]

SivarajKK, et al. . Regional specialization and fate specification of bone stromal cells in skeletal development. Cell Rep., 2021, 36: 109352.

[216]

KimJM, et al. . DJ-1 promotes angiogenesis and osteogenesis by activating FGF receptor-1 signaling. Nat. Commun., 2012, 3. 1296

[217]

ZhangL, et al. . Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion. Stem Cell Res. Ther., 2020, 11: 38.

[218]

LiuH, et al. . Endothelial progenitor cells improve the therapeutic effect of mesenchymal stem cell sheets on irradiated bone defect repair in a rat model. J. Transl. Med., 2018, 16. 137

[219]

WuJ, et al. . Endothelial cell-derived lactate triggers bone mesenchymal stem cell histone lactylation to attenuate osteoporosis. Adv. Sci., 2023, 10: e2301300.

[220]

LiJ, et al. . Type H vessel/platelet-derived growth factor receptor β+ perivascular cell disintegration is involved in vascular injury and bone loss in radiation-induced bone damage. Cell Prolif., 2023, 56: e13406.

[221]

JamesAW, et al. . Pericytes for the treatment of orthopedic conditions. Pharmacol. Ther., 2017, 171: 93-103.

[222]

IssabekovaA, et al. . The therapeutic potential of pericytes in bone tissue regeneration. Biomedicines, 2023, 12: 21.

[223]

GeevargheseA, HermanIM. Pericyte-endothelial crosstalk: implications and opportunities for advanced cellular therapies. Transl. Res., 2014, 163: 296-306.

[224]

DimitriouR, JonesE, McGonagleD, GiannoudisPV. Bone regeneration: current concepts and future directions. BMC Med., 2011, 9. 66

[225]

Uhthoff, H. K. & Rahn, B. A. Healing patterns of metaphyseal fractures. Clin. Orthop Relat. Res.160, 295–303 (1981).

[226]

ThompsonZ, MiclauT, HuD, HelmsJA. A model for intramembranous ossification during fracture healing. J. Orthop. Res., 2002, 20: 1091-1098.

[227]

AspenbergP, SandbergO. Distal radial fractures heal by direct woven bone formation. Acta Orthop., 2013, 84: 297-300.

[228]

LoiF, et al. . Inflammation, fracture and bone repair. Bone, 2016, 86: 119-130.

[229]

QiuP, et al. . Periosteal matrix-derived hydrogel promotes bone repair through an early immune regulation coupled with enhanced angio- and osteogenesis. Biomaterials, 2020, 227: 119552.

[230]

MarenzanaM, ArnettTR. The key role of the blood supply to bone. Bone Res., 2013, 1: 203-215.

[231]

PhillipsAM. Overview of the fracture healing cascade. Injury, 2005, 36: S5-S7.

[232]

PajarinenJ, et al. . Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials, 2019, 196: 80-89.

[233]

KimYW, et al. . Integration of single-cell transcriptomes and biological function reveals distinct behavioral patterns in bone marrow endothelium. Nat. Commun., 2022, 13. 7235

[234]

DohleE, et al. . Macrophage-mediated angiogenic activation of outgrowth endothelial cells in co-culture with primary osteoblasts. Eur. Cell Mater., 2014, 27: 149-164. discussion 164-145

[235]

GuL, et al. . Atf4 regulates angiogenic differences between alveolar bone and long bone macrophages by regulating M1 polarization, based on single-cell RNA sequencing, RNA-seq and ATAC-seq analysis. J. Transl. Med., 2023, 21. 193

[236]

SmithRJJr., YiT, NasiriB, BreuerCK, AndreadisST. Implantation of VEGF-functionalized cell-free vascular grafts: regenerative and immunological response. FASEB J., 2019, 33: 5089-5100.

[237]

LiX, DaiY, ShenT, GaoC. Induced migration of endothelial cells into 3D scaffolds by chemoattractants secreted by pro-inflammatory macrophages in situ. Regen. Biomater., 2017, 4: 139-148.

[238]

ZhangY, et al. . Tailoring surface stiffness to modulate senescent macrophage immunomodulation: Implications for osteo-/angio-genesis in aged bone regeneration. Biomater. Adv., 2024, 165: 214010.

[239]

GuoL, et al. . CD163+ macrophages promote angiogenesis and vascular permeability accompanied by inflammation in atherosclerosis. J. Clin. Invest., 2018, 128: 1106-1124.

[240]

WangY, et al. . M2 macrophages promote vasculogenesis during retinal neovascularization by regulating bone marrow-derived cells via SDF-1/VEGF. Cell Tissue Res., 2020, 380: 469-486.

[241]

LinW, et al. . Endothelial exosomes work as a functional mediator to activate macrophages. Front. Immunol., 2023, 14: 1169471.

[242]

HeS, et al. . Endothelial extracellular vesicles modulate the macrophage phenotype: potential implications in atherosclerosis. Scand. J. Immunol., 2018, 87: e12648.

[243]

ChenY, et al. . Exosomal Lnc NEAT1 from endothelial cells promote bone regeneration by regulating macrophage polarization via DDX3X/NLRP3 axis. J. Nanobiotechnol., 2023, 21. 98

[244]

ChenL, et al. . Multifunctional hydrogel enhances bone regeneration through sustained release of stromal cell-derived factor-1alpha and exosomes. Bioact. Mater., 2023, 25: 460-471

[245]

WangJ, et al. . Break monopoly of polarization: CD301b+ macrophages play positive roles in osteoinduction of calcium phosphate ceramics. Appl. Mater. Today, 2021, 24: 101111.

[246]

WangJ, et al. . CD301b+ macrophages mediate angiogenesis of calcium phosphate bioceramics by CaN/NFATc1/VEGF axis. Bioact. Mater., 2022, 15: 446-455

[247]

WangC, et al. . CD301b+ macrophage: the new booster for activating bone regeneration in periodontitis treatment. Int. J. Oral. Sci., 2023, 15: 19.

[248]

ChampagneCM, TakebeJ, OffenbacherS, CooperLF. Macrophage cell lines produce osteoinductive signals that include bone morphogenetic protein-2. Bone, 2002, 30: 26-31.

[249]

LuLY, et al. . Pro-inflammatory M1 macrophages promote osteogenesis by mesenchymal stem cells via the COX-2-prostaglandin E2 pathway. J. Orthop. Res., 2017, 35: 2378-2385.

[250]

HuangY, et al. . Bone marrow mesenchymal stem cell-derived exosomes promote rotator cuff tendon-bone healing by promoting angiogenesis and regulating M1 macrophages in rats. Stem Cell Res. Ther., 2020, 11: 496.

[251]

QiY, et al. . M1 macrophage-derived exosomes transfer miR-222 to induce bone marrow mesenchymal stem cell apoptosis. Lab. Investig., 2021, 101: 1318-1326.

[252]

BastianO, et al. . Systemic inflammation and fracture healing. J. Leukoc. Biol., 2011, 89: 669-673.

[253]

ClaesL, RecknagelS, IgnatiusA. Fracture healing under healthy and inflammatory conditions. Nat. Rev. Rheumatol., 2012, 8: 133-143.

[254]

Romero-LopezM, et al. . Macrophage effects on mesenchymal stem cell osteogenesis in a three-dimensional in vitro bone model. Tissue Eng. Part A, 2020, 26: 1099-1111.

[255]

GongL, ZhaoY, ZhangY, RuanZ. The macrophage polarization regulates MSC osteoblast differentiation in vitro. Ann. Clin. Lab. Sci., 2016, 46: 65-71

[256]

MahonOR, et al. . Nano-particle-mediated M2 macrophage polarization enhances bone formation and MSC osteogenesis in an IL-10 dependent manner. Biomaterials, 2020, 239: 119833.

[257]

PuP, et al. . Mechanical force induces macrophage-derived exosomal UCHL3 promoting bone marrow mesenchymal stem cell osteogenesis by targeting SMAD1. J. Nanobiotechnol., 2023, 21. 88

[258]

LiangZY, et al. . Mesenchymal stem cell-derived exosomal MiRNAs promote M2 macrophages polarization: therapeutic opportunities for spinal cord injury. Front. Mol. Neurosci., 2022, 15: 926928.

[259]

BixelMG, et al. . Angiogenesis is uncoupled from osteogenesis during calvarial bone regeneration. Nat. Commun., 2024, 15. 4575

[260]

DiomedeF, et al. . Functional relationship between osteogenesis and angiogenesis in tissue regeneration. Int. J. Mol. Sci., 2020, 21: 3242.

[261]

BurgerMG, et al. . Robust coupling of angiogenesis and osteogenesis by VEGF-decorated matrices for bone regeneration. Acta Biomater., 2022, 149: 111-125.

[262]

LiuX, ZhangP, GuY, GuoQ, LiuY. Type H vessels: functions in bone development and diseases. Front. Cell Dev. Biol., 2023, 11: 1236545.

[263]

RuanZ, et al. . Metformin accelerates bone fracture healing by promoting type H vessel formation through inhibition of YAP1/TAZ expression. Bone Res., 2023, 11: 45.

[264]

ShaoJ, et al. . A dual role of HIF1α in regulating osteogenesis-angiogenesis coupling. Stem Cell Res. Ther., 2022, 13: 59.

[265]

GuanH, et al. . Magnetic aggregation-induced bone-targeting nanocarrier with effects of Piezo1 activation and osteogenic-angiogenic coupling for osteoporotic bone repair. Adv. Mater., 2024, 36: e2312081.

[266]

ZhouC, HuG, LiY, ZhengS. Polydatin accelerates osteoporotic bone repair by inducing the osteogenesis-angiogenesis coupling of bone marrow mesenchymal stem cells via the PI3K/AKT/GSK- 3β/β-catenin pathway. Int. J. Surg., 2024, 111: 411-425.

[267]

CaiX, YaoZ, LiL, HuangJ. Role of DKK4 in tumorigenesis and tumor progression. Int. J. Biol. Sci., 2018, 14: 616-621.

[268]

ZhengS, HuG, ZhengJ, LiY, LiJ. Osthole accelerates osteoporotic fracture healing by inducing the osteogenesis-angiogenesis coupling of BMSCs via the Wnt/β-catenin pathway. Phytother. Res., 2024, 38: 4022-4035.

[269]

HuY, et al. . Defactinib attenuates osteoarthritis by inhibiting positive feedback loop between H-type vessels and MSCs in subchondral bone. J. Orthop. Transl., 2020, 24: 12-22

[270]

ZhangD, et al. . Single-cell RNA sequencing reveals neurovascular-osteochondral network crosstalk during temporomandibular joint osteoarthritis: Pilot study in a human condylar cartilage. Heliyon, 2023, 9: e20749.

[271]

NagaoM, et al. . Vascular endothelial growth factor in cartilage development and osteoarthritis. Sci. Rep., 2017, 7. 13027

[272]

LiuZ, et al. . Selective formation of osteogenic and vasculogenic tissues for cartilage regeneration. Adv. Healthc. Mater., 2023, 12: e2202008.

[273]

WuZ, et al. . Antioxidant PDA-PEG nanoparticles alleviate early osteoarthritis by inhibiting osteoclastogenesis and angiogenesis in subchondral bone. J. Nanobiotechnol., 2022, 20. 479

[274]

KimHJ, et al. . Zoledronate enhances osteocyte-mediated osteoclast differentiation by IL-6/RANKL Axis. Int. J. Mol. Sci., 2019, 20: 1467.

[275]

TsengHC, et al. . Bisphosphonate-induced differential modulation of immune cell function in gingiva and bone marrow in vivo: role in osteoclast-mediated NK cell activation. Oncotarget, 2015, 6: 20002-20025.

[276]

XuJJ, ZhouJ, WangSL. [Osteoradionecrosis and bisphosphonate-related osteonecrosis of the jaw: a review of the pathogenesis and treatment]. Zhonghua Kou Qiang Yi Xue Za Zhi, 2021, 56: 404-409

[277]

WatanabeJ, et al. . Extracellular vesicles of stem cells to prevent BRONJ. J. Dent. Res., 2020, 99: 552-560.

[278]

ChenH, LiuZ, YueK, DingJ, HeY. Immune microenvironment: novel perspectives on bone regeneration disorder in osteoradionecrosis of the jaws. Cell Tissue Res., 2023, 392: 413-430.

[279]

DeshpandeSS, et al. . Stem cells rejuvenate radiation-impaired vasculogenesis in murine distraction osteogenesis. Plast. Reconstruct. Surg., 2015, 135: 799-806.

[280]

MaT, et al. . Research progress in the pathogenesis of hormone-induced femoral head necrosis based on microvessels: a systematic review. J. Orthop. Surg. Res., 2024, 19. 265

[281]

ShanH, et al. . Effects of astragaloside IV on glucocorticoid-induced avascular necrosis of the femoral head via regulating Akt-related pathways. Cell Prolif., 2023, 56: e13485.

[282]

PengP, WangX, QiuC, ZhengW, ZhangH. Extracellular vesicles from human umbilical cord mesenchymal stem cells prevent steroid-induced avascular necrosis of the femoral head via the PI3K/AKT pathway. Food Chem. Toxicol., 2023, 180: 114004.

[283]

ChenJ, et al. . Human umbilical cord mesenchymal stem cells promote steroid-induced osteonecrosis of the femoral head repair by improving microvascular endothelial cell function. Aging, 2024, 16: 7928-7945

[284]

XuH, et al. . Cotransplantation of mesenchymal stem cells and endothelial progenitor cells for treating steroid-induced osteonecrosis of the femoral head. Stem Cells Transl. Med., 2021, 10: 781-796.

[285]

CheahE, WuZ, ThakurSS, O’CarrollSJ, SvirskisD. Externally triggered release of growth factors—a tissue regeneration approach. J. Control Release, 2021, 332: 74-95.

[286]

SubbiahR, et al. . Triple growth factor delivery promotes functional bone regeneration following composite musculoskeletal trauma. Acta Biomater., 2021, 127: 180-192.

[287]

KrishnanL, et al. . Delivery vehicle effects on bone regeneration and heterotopic ossification induced by high dose BMP-2. Acta Biomater., 2017, 49: 101-112.

[288]

WuF, et al. . Discoidin domain receptor 2 is an important modulator of BMP signaling during heterotopic bone formation. Bone Res., 2025, 13: 7.

[289]

XiongY, et al. . miRNA-26a-5p accelerates healing via downregulation of PTEN in fracture patients with traumatic brain injury. Mol. Ther. Nucleic Acids, 2019, 17: 223-234.

[290]

NovakS, et al. . Modulation of Notch1 signaling regulates bone fracture healing. J. Orthop. Res., 2020, 38: 2350-2361.

[291]

NovakS, et al. . Endothelial to mesenchymal Notch signaling regulates skeletal repair. JCI Insight, 2024, 9: e181073.

[292]

RemarkLH, et al. . Loss of Notch signaling in skeletal stem cells enhances bone formation with aging. Bone Res., 2023, 11: 50.

[293]

HuJ, et al. . Genotypic and phenotypic spectrum and pathogenesis of WNT1 variants in a large cohort of patients with OI/osteoporosis. J. Clin. Endocrinol. Metab., 2023, 108: 1776-1786.

[294]

FanH, et al. . Electroacupuncture stimulation at CV4 prevents ovariectomy-induced osteoporosis in rats via Wnt-beta-catenin signaling. Mol. Med. Rep., 2016, 13: 2485-2491.

[295]

WuF, et al. . Exosomes derived from BMSCs in osteogenic differentiation promote type H blood vessel angiogenesis through miR-150-5p mediated metabolic reprogramming of endothelial cells. Cell Mol. Life Sci., 2024, 81: 344.

[296]

ChenH, et al. . Urchin-like ceria nanoparticles for enhanced gene therapy of osteoarthritis. Sci. Adv., 2023, 9: eadf0988.

[297]

WuY, LuX, ShenB, ZengY. The therapeutic potential and role of miRNA, lncRNA, and circRNA in osteoarthritis. Curr. Gene Ther., 2019, 19: 255-263.

[298]

XuX, et al. . Histone modification of osteogenesis-related genes triggered by substrate topography promotes human mesenchymal stem cell differentiation. ACS Appl. Mater. interfaces, 2023, 15: 29752-29766.

[299]

WangZ, et al. . Epigenetics: novel crucial approach for osteogenesis of mesenchymal stem cells. J. Tissue Eng., 2023, 14: 20417314231175364.

[300]

KumarA, et al. . Manufacturing functional hydrogels for inducing angiogenic-osteogenic coupled progressions in hard tissue repairs: prospects and challenges. Biomater. Sci., 2022, 10: 5472-5497.

[301]

ZhaoH, et al. . 3D printing of dual-cell delivery titanium alloy scaffolds for improving osseointegration through enhancing angiogenesis and osteogenesis. BMC Musculoskelet. Disord., 2021, 22. 734

[302]

BayerEA, et al. . (*) Programmed platelet-derived growth factor-bb and bone morphogenetic protein-2 delivery from a hybrid calcium phosphate/alginate scaffold. Tissue Eng. Part A, 2017, 23: 1382-1393.

[303]

SongC, LiuR, FangY, GuH, WangY. Developing functional hydrogels for treatment of oral diseases. Smart Med., 2024, 3: e20240020.

[304]

YanR, et al. . Trace element-augmented titanium implant with targeted angiogenesis and enhanced osseointegration in osteoporotic rats. Front. Chem., 2022, 10: 839062.

[305]

WenZ, et al. . Mesoporous TiO(2) coatings regulate ZnO nanoparticle loading and Zn(2+) release on titanium dental implants for sustained osteogenic and antibacterial activity. ACS Appl. Mater. Interfaces, 2023, 15: 15235-15249.

[306]

WangX, TangM. Bioceramic materials with ion-mediated multifunctionality for wound healing. Smart Med., 2022, 1: e20220032.

[307]

DengZ, et al. . Hypoxia-mimicking cobalt-doped borosilicate bioactive glass scaffolds with enhanced angiogenic and osteogenic capacity for bone regeneration. Int. J. Biol. Sci., 2019, 15: 1113-1124.

[308]

JinYZ, LeeJH. Mesenchymal stem cell therapy for bone regeneration. Clin. Orthop. Surg., 2018, 10: 271-278.

[309]

HuangS, XuL, SunY, ZhangY, LiG. The fate of systemically administrated allogeneic mesenchymal stem cells in mouse femoral fracture healing. Stem Cell Res. Ther., 2015, 6: 206.

[310]

LeiF, et al. . Treatment of inflammatory bone loss in periodontitis by stem cell-derived exosomes. Acta Biomater., 2022, 141: 333-343.

[311]

LuoY, et al. . Ginsenoside RG1 enhances the paracrine effects of bone marrow-derived mesenchymal stem cells on radiation-induced intestinal injury. Aging, 2020, 13: 1132-1152.

[312]

LiuS, et al. . An osteoimmunomodulatory biopatch potentiates stem cell therapies for bone regeneration by simultaneously regulating IL-17/ferroptosis signaling pathways. Adv. Sci., 2024, 11: e2401882.

[313]

WangX, et al. . The optimal time to inject bone mesenchymal stem cells for fracture healing in a murine model. Stem Cell Res. Ther., 2018, 9: 272.

[314]

Zigdon-GiladiH, BickT, LewinsonD, MachteiEE. Co-transplantation of endothelial progenitor cells and mesenchymal stem cells promote neovascularization and bone regeneration. Clin. Implant Dent. Relat. Res., 2015, 17: 353-359.

[315]

JiaY, ZhangC, ZhengX, GaoM. Co-cultivation of progenitor cells enhanced osteogenic gene expression and angiogenesis potential in vitro. J. Int. Med. Res., 2021, 49: 3000605211004024.

[316]

BoulandC, et al. . Cross-talk between mesenchymal stromal cells (MSCs) and endothelial progenitor cells (EPCs) in bone regeneration. Front. Cell Dev. Biol., 2021, 9: 674084.

[317]

WangH, et al. . Construction of vascularized tissue-engineered bone with nHA-coated BCP bioceramics loaded with peripheral blood-derived MSC and EPC to repair large segmental femoral bone defect. ACS Appl. Mater. Interfaces, 2023, 15: 249-264.

[318]

GengZ, et al. . Novel bionic topography with MiR-21 coating for improving bone-implant integration through regulating cell adhesion and angiogenesis. Nano Lett., 2020, 20: 7716-7721.

[319]

HeWZ, et al. . miR-188-3p targets skeletal endothelium coupling of angiogenesis and osteogenesis during ageing. Cell Death Dis., 2022, 13: 494.

[320]

HanY, YouX, XingW, ZhangZ, ZouW. Paracrine and endocrine actions of bone-the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res., 2018, 6: 16.

[321]

WangZ, ZhaoZ, GaoB, ZhangL. Exosome mediated biological functions within skeletal microenvironment. Front. Bioeng. Biotechnol., 2022, 10: 953916.

[322]

TaoSC, GuoSC. Extracellular vesicles in bone: “dogrobbers” in the “eternal battle field”. Cell Commun. Signal., 2019, 17: 6.

[323]

ZappalaA, et al. . Functional roles of connexins and gap junctions in osteo-chondral cellular components. Int. J. Mol. Sci., 2023, 24: 4156.

[324]

ZhuS, et al. . Coupling factors and exosomal packaging microRNAs involved in the regulation of bone remodelling. Biol. Rev. Camb. Philos. Soc., 2018, 93: 469-480.

Funding

National Natural Science Foundations of China 82230030 and 81871492, Beijing International Science and Technology Cooperation Project Z221100002722003, Beijing Natural Science Foundation L234017, Peking University Clinical Medicine Plus X – Young Scholars Project PKU2023LCXQ004, Ten-Thousand Talents Program QNBJ2019-2, Key R & D Plan of Ningxia Hui Autonomous Region 2020BCG01001.

RIGHTS & PERMISSIONS

The Author(s)

AI Summary AI Mindmap
PDF

365

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/