Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
March-2026 Volume 57 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
March-2026 Volume 57 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)

  • Authors:
    • Ruo-Mei Cui
    • Mai Zheng
    • Jian-Bin Hong
    • Zheng-Xiang Wang
    • Yu-Fang Cun
    • Shu-Ji Gao
    • Yan-Lin Zhu
    • Zi-Bin Yang
    • Ming-Wei Liu
  • View Affiliations / Copyright

    Affiliations: Department of Rheumatology, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China, Department of Orthopedics, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China, Department of Spinal Surgery, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China, Department of Pharmacy, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China, Emergency Department, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China, Emergency Department, Dali Bai Autonomous Prefecture People's Hospital, Dali, Yunnan 671000, P.R. China
    Copyright: © Cui et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 67
    |
    Published online on: January 21, 2026
       https://doi.org/10.3892/ijmm.2026.5738
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:


Abstract

Musculoskeletal crosstalk is essential for maintaining the balance of bone metabolism, with macrophage‑derived exosomes emerging as key regulators of this process. Exosomes, small extracellular vesicles secreted by cells, carry a variety of bioactive molecules; proteins, lipids, mRNAs and miRNAs and facilitate intercellular communication by transferring these cargos to recipient cells. Specifically, macrophage‑derived exosomes mediate muscle‑bone interactions by transferring key regulators such as insulin‑like growth factor‑1 (IGF‑1) and fibroblast growth factor‑2 (FGF‑2), thereby playing a pivotal role in bone metabolic homeostasis. Macrophages are classified into pro‑inflammatory M1 and anti‑inflammatory M2 phenotypes, each performing distinct functions in immune responses. Exosomes from M1 macrophages typically carry pro‑inflammatory factors that can activate osteoclastic bone resorption, disrupting bone metabolism in pathological conditions. By contrast, exosomes from M2 macrophages often contain anti‑inflammatory factors that promote tissue repair and bone formation. In the context of bone metabolism, exosomes from M1 and M2 macrophages modulate muscle‑bone signaling by delivering regulators that influence the expression of IGF‑1 and FGF‑2, affecting osteoblast proliferation, differentiation, and mineralization. M1 macrophage‑derived exosomes activate signaling pathways such as NF‑κB and MAPK through the transfer of pro‑inflammatory cargo, thereby enhancing bone resorption. By contrast, exosomes from M2 macrophages can suppress pro‑inflammatory signaling while activating pathways like TGF‑β and PI3K/Akt, promoting bone synthesis and repair. As critical myokines, IGF‑1 and FGF‑2 not only support muscle growth, repair, and maintenance but also directly influence bone remodeling through musculoskeletal crosstalk.

Introduction

Musculoskeletal crosstalk is a fundamental physiological process that maintains human health, involving the dynamic interaction and regulation between bone and muscle through intricate molecular signaling mechanisms (1). Myokines such as insulin-like growth factor-1 (IGF-1) and fibroblast growth factor-2 (FGF-2) are pivotal in mediating this crosstalk (2). IGF-1 regulates muscle growth and metabolism by promoting myocyte proliferation and repair, while FGF-2 plays a critical role in muscle regeneration and repair (3). Notably, both IGF-1 and FGF-2 not only regulate muscle growth but also markedly influence bone metabolism, bone density maintenance and the repair and regeneration of bone tissue (4). This inter-system regulatory mechanism highlights the importance of musculoskeletal crosstalk as a critical area of research (5). Recent developments in intercellular communication have identified macrophage-derived exosomes as emerging signaling mediators, gaining increasing attention in the scientific community (6). Exosomes are small extracellular vesicles secreted by cells that carry a range of bioactive molecules, including proteins, RNAs, and lipids, facilitating intercellular communication and the regulation of cellular functions and tissue physiology (7).

Macrophages regulate musculoskeletal interactions through the secretion of exosomes (8). Exosomes derived from M1 and M2 macrophages exhibit distinct effects on musculoskeletal metabolism (Tables I and II) (9-13). M1 macrophages, which are pro-inflammatory, release exosomes enriched with pro-inflammatory cytokines that can stimulate osteoclastic bone resorption, potentially contributing to osteoporosis and bone damage. By contrast, M2 macrophages, which possess anti-inflammatory properties, secrete exosomes containing anti-inflammatory factors that promote bone formation and repair. Macrophages modulate muscle-bone signaling through the secretion of specific exosome subtypes. Exosome-associated factors like IGF-1 and FGF-2 regulate bone metabolic homeostasis, either promoting or inhibiting bone remodeling.

Table I

Comparative analysis of M1 vs. M2 macrophage-derived exosomes in bone metabolism regulation.

Table I

Comparative analysis of M1 vs. M2 macrophage-derived exosomes in bone metabolism regulation.

ParameterM1-ExosM2-Exos
BiogenesisFrom LPS/IFN-γ-polarized macrophagesProduced by IL-4/IL-13-stimulated macrophages
Surface MarkersCD86+, iNOS+, MHC-II+ CD206+, Arg-1+, CD163+
Key cargosmiRNAs: miR-155, let-7c
Proteins: TNF-α, IL-6, IL-1β
miRNAs: miR-223, miR-146a
Proteins: IL-10, TGF-β, CCL18
Osteoblast effectsInhibits differentiation via Wnt/β-catenin suppression ALP activity↓Promotes mineralization via BMP/Runx2 pathway COL1A1 and OCN expression ↑
Osteoclast effectsEnhances RANKL-induced differentiation TRAP+ multinucleated cells↑Secretes OPG to inhibit RANKL Resorption pit area ↓ (by 40-60%)
Metabolic reprogrammingGlycolysis↑ (HK2/LDHA↑) ROS production↑Oxidative phosphorylation↑ (ATP5A1↑) FAO via PPARγ activation
Therapeutic potentialAcute bone injury (such as fracture hematoma clearance)Chronic conditions (such as osteoporosis, non-union fractures)
(Refs.)(9,10)(11)

[i] Upward arrow (↑) indicates increase; downward arrow (↓) indicates decrease. M1-Exos, M1 macrophage-derived exosomes; M2-Exos, M2 macrophage-derived exosomes; LPS, lipopolysaccharide; IFN-γ, interferon γ; iNOS+, inducible nitric oxide synthase; MHC-II, major histocompatibility complex class II; miRNAs/miRs, microRNAs; ALP, alkaline phosphatase; OPG, osteoprotegerin; TGF-β, transforming growth factor β; ROS, reactive oxygen species; BMP, bone morphogenetic protein; OCN, osteocalcin; RANKL, receptor activator of nuclear factor-κB ligand; HK2, hexokinase 2; LDHA, lactate dehydrogenase A; ATP5A1, ATP synthase-alpha; PPARγ, peroxisome proliferator-activated receptor gamma; FAO, fatty acid oxidation; RUNX2, Runt-related transcription factor 2.

Table II

Comparative analysis of M1 and M2 macrophage-derived exosomes in muscle metabolism regulation.

Table II

Comparative analysis of M1 and M2 macrophage-derived exosomes in muscle metabolism regulation.

ParameterM1-ExosM2-Exos
Polarization signalInduced by LPS/IFN-γInduced by IL-4/IL-13
Key cargosmiRNAs: miR-155, miR-27a
Proteins: TNF-α, IL-6, IL-1β
miRNAs: miR-223, miR-206
Proteins: IL-10, IGF-1, TGF-β
MyogenesisInhibits satellite cell differentiation (MyoD↓)
Promotes atrophy (MuRF-1↑)
Enhances myoblast fusion (MyoG↑)
Reduces fibrosis (Collagen III↓)
Glucose metabolismInduces insulin resistance (IRS-1 phosphorylation↓)
GLUT4 translocation↓
Improves insulin sensitivity (Akt activation↑)
Glycogen synthesis↑
Lipid metabolismPromotes lipolysis (ATGL↑)
Mitochondrial dysfunction (ROS↑)
Enhances fatty acid oxidation (CPT1B↑)
Mitochondrial biogenesis (PGC-1α↑)
Clinical relevanceChronic inflammation (such as sarcopenia, DMD)Muscle regeneration (such as injury, aging)
(Refs.)(12)(13)

[i] Upward arrow (↑) indicates increase; downward arrow (↓) indicates decrease. M1-Exos, M1 macrophage-derived exosomes; M2-Exos, M2 macrophage-derived exosomes; LPS, lipopolysaccharide; IGF1, insulin-like growth factor 1; TGF-β, transforming growth factor beta; MyoD, myogenic differentiation 1; MuRF1, muscle ring finger-1; MYOG, human myogenin; IRS1, insulin receptor substrate 1; Akt, protein kinase B; GLUT4, Glucose transporter type-4; ROS, reactive oxygen species; CPT1B, Carnitine palmitoyltransferase 1b; PGC-1α, peroxisome proliferator-activated receptor-gamma coactivator 1α; DMD, Duchenne muscular dystrophy.

The present review aimed to synthesize recent insights into macrophage-derived exosomes and their role in regulating musculoskeletal crosstalk, with a focus on their involvement in IGF-1- and FGF-2-mediated signaling pathways. It examined how exosomes influence the functions of these key factors in bone metabolism, elucidated their roles in muscle and bone homeostasis and suggested future research avenues. Additionally, it sought to provide a theoretical framework for the development of therapeutic strategies targeting exosomes, particularly for the treatment of bone metabolic disorders and musculoskeletal diseases (14).

The importance of muscle factors IGF-1 and FGF-2 in musculoskeletal crosstalk

Within the musculoskeletal system, muscle-derived factors such as IGF-1 and FGF-2 are essential in mediating tissue crosstalk (Table III) (15-18). These factors coordinate the development and repair of both bone and muscle by regulating their bidirectional interactions. IGF-1 is a pivotal growth factor that promotes muscle cell proliferation and differentiation while also exerting significant regulatory effects on the skeletal system (19). Binding to its receptor activates the PI3K/Akt pathway, fostering muscle growth and enhancing bone mineralization and density, thereby serving as a critical link between bone and muscle (20). FGF-2 is a key regulator of fibroblasts and bone marrow-derived cells, playing an indispensable role in bone repair and muscle regeneration (21). Upon binding to fibroblast growth factor receptors (FGFRs), FGF-2 activates downstream pathways, including MAPK/ERK and PI3K/Akt, that promote the growth and differentiation of osteoblasts and myocytes (22). Notably, FGF-2 supports effective repair of muscle and bone tissues following exercise or trauma.

Table III

Comparative roles of IGF-1 and FGF-2 in muscle and bone metabolism.

Table III

Comparative roles of IGF-1 and FGF-2 in muscle and bone metabolism.

CategoryIGF-1FGF-2
Primary sourceLiver (endocrine), muscle/osteoblasts (paracrine)Mesenchymal stem cells, osteoblasts, damaged tissues
Muscle metabolismAnabolic Effects:
Activates PI3K/Akt/mTOR → protein synthesis↑
Proliferative Effects:
Binds FGFR1→MAPK → myoblast proliferation↑
Suppresses FoxO/MuRF-1 → atrophy↓
Enhances satellite cell differentiation
Synergizes with IGF-1 for regeneration
Reduces fibrosis (TGF-β inhibition)
Bone metabolism Pro-osteogenic:
Stimulates Runx2/Osterix → mineralization↑
Inhibits RANKL → osteoclastogenesis↓
ECM production (Collagen I↑)
Biphasic Action:
Low dose: BMP-2 synergy → osteogenesis↑
High dose: Angiogenesis (VEGF/FGFR1)→ bone remodeling
Metabolic influenceGLUT4 translocation↑ → glucose uptake↑
PPARδ activation → fatty acid oxidation↑
Glycolysis↑ via HIF-1α
Modulates bone-vascular coupling (PDGF-BB interaction)
Receptor pathway IGF-1R→IRS-1→PI3K/AktFGFR1-4→RAS/MAPK or PLCγ
Therapeutic userhIGF-1 for sarcopenia
Bone defect scaffolds (sustained release)
FGF-2-coated implants
Muscle injury repair (clinical trials)
(Refs.)(15,16)(17,18)

[i] Upward arrow (↑) indicates increase; downward arrow (↓) indicates decrease. IGF-1, Insulin-like growth factor-1; FGF-2, Fibroblast growth factor-2; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; FoxO, Forkhead box O; MuRF-1, muscle RING finger-1; FGFR, Fibroblast growth factor receptor; MAPK, Mitogen-activated protein kinase; TGF-β, Transforming growth factor beta; Runx2, Runt-related transcription factor 2; RANKL, Receptor activator of nuclear factor kappa-B ligand; ECM, Extracellular matrix; BMP-2, Bone morphogenetic protein-2; VEGF, Vascular endothelial growth factor; GLUT4, glucose transporter type 4; PPARδ, Peroxisome proliferator-activated receptor delta; HIF-1α, hypoxia-inducible factor 1-alpha; PDGF-BB, Platelet-derived growth factor BB; RAS, rat sarcoma; PLCγ, phospholipase C gamma; rhIGF-1, recombinant human IGF-1.

Musculoskeletal crosstalk molecular signaling pathways

The physiological connection between muscle and bone
The role of mechanical signal transduction in musculoskeletal interaction

Mechanical signal transduction is crucial in musculoskeletal interactions (23). The close relationship between bone and muscle during exercise is particularly dependent on mechanical stimulation (Fig. 1) (23,24). During exercise, muscles generate mechanical tension on bones, stimulating bone growth and remodeling, while also modulating various aspects of bone metabolism (25). For instance, during weight-bearing exercise, muscle contraction generates mechanical loads, such as pressure and traction, on bone. These forces are transmitted via mechanoreceptors on bone cells, regulating the balance between bone resorption and formation (26). In response to mechanical stimuli, bone activates several signaling pathways, including mechanosensory pathways such as the Yes-associated protein/transcriptional coactivator with PDZ-binding motif (YAP/TAZ) pathway, which activates osteoblasts and other bone-related cells to promote bone mineralization (27). Mechanical signals also regulate muscle growth (28). Muscle adapts to mechanical loads through signaling molecules such as growth factors and protein kinases, which promote muscle fiber growth and proliferation (29). This feedback mechanism fosters muscle hyperplasia while simultaneously influencing bone, facilitating integrated musculoskeletal crosstalk and highlighting the interdependence of bone and muscle throughout human development (30).

Regulatory mechanism of mechanical
signal transduction on the musculoskeletal system. Mechanical
stress inhibits the Hippo pathway, activating Wnt/β-catenin
signaling. It also suppresses sclerostin, further promoting Wnt
pathway activation. Additionally, mechanical stress regulates
integrins to activate the FAK and RhoA/ROCK pathways, stimulating
osteoblasts and other bone-related cells, thus promoting bone
mineralization. Furthermore, mechanical stress regulates muscle
growth through activation of AMPK, PI3K/Akt/mTORC1, and
mitochondrial metabolism-related pathways. FAK, Focal adhesion
kinase; RhoA, Ras homolog gene family member A; ROCK,
Rho-associated kinase; AMPK, Adenosine monophosphate-activated
protein kinase; PI3K, phosphatidylinositol-3-kinase; Akt, protein
kinase B; mTORC1, mammalian target of rapamycin complex 1; PGC-1α,
Peroxisome proliferators-activated receptor γ coactivator 1α.

Figure 1

Regulatory mechanism of mechanical signal transduction on the musculoskeletal system. Mechanical stress inhibits the Hippo pathway, activating Wnt/β-catenin signaling. It also suppresses sclerostin, further promoting Wnt pathway activation. Additionally, mechanical stress regulates integrins to activate the FAK and RhoA/ROCK pathways, stimulating osteoblasts and other bone-related cells, thus promoting bone mineralization. Furthermore, mechanical stress regulates muscle growth through activation of AMPK, PI3K/Akt/mTORC1, and mitochondrial metabolism-related pathways. FAK, Focal adhesion kinase; RhoA, Ras homolog gene family member A; ROCK, Rho-associated kinase; AMPK, Adenosine monophosphate-activated protein kinase; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; mTORC1, mammalian target of rapamycin complex 1; PGC-1α, Peroxisome proliferators-activated receptor γ coactivator 1α.

The role of endocrine factors in musculoskeletal communication

Beyond mechanical signals, endocrine factors also play a vital role in musculoskeletal crosstalk (31). Muscles and bones mutually regulate and support each other via various endocrine factors, including IGF-1, growth hormone, sex hormones and FGF-2 (Fig. 2) (32). During growth, IGF-1 secreted by muscles promotes muscle development and, via circulation, also affects the skeletal system, facilitating bone formation (33). Binding to the IGF-1 receptor (IGF-1R) in bone activates the PI3K-Akt and MAPK pathways, regulating osteoblast proliferation and differentiation (34). Sex hormones, particularly estrogen, are critical in regulating muscle-bone interactions (35). Estrogen maintains bone density and mass by promoting bone formation and inhibiting resorption, while also enhancing musculoskeletal synergy by regulating muscle strength and endurance (Fig. 3) (35). FGF-2, as an endocrine factor in bone, contributes to bone repair and regulates growth and mineralization by binding to FGFRs (Fig. 2) (36). Particularly during aging or following fracture, FGF-2 supports bone repair by stimulating osteoblast proliferation and differentiation (37).

Regulatory pathways of muscle factors
on the musculoskeletal system. IGF-1 modulates muscle metabolism
via the PI3K/Akt/mTOR pathway and bone metabolism through PI3K/Akt
and MAPK/ERK signaling. FGF-2 regulates muscle metabolism via
MAPK/ERK and bone metabolism through BMP-2/Smad signaling.
Myostatin influences muscle metabolism via Smad3 and P21, and bone
metabolism through Smad2 and Wnt/β-catenin pathways. PGC-1α
stimulates PNDC5, promoting Irisin release from muscle; Irisin
subsequently regulates bone metabolism via P38MAPK/ERK and
RANKL/NFATc1 signaling. IGF-1, Insulin-like growth factor-1; FGF-2,
Fibroblast growth factor 2; GSK3, Glycogen Synthase Kinase-3;
IGFBP-3, insulin-like growth factor binding protein 3; IGFBP-5,
insulin-like growth factor binding protein 5; MyoD, Myoblast
determination protein 1; PNDC, peroxynitrite decomposition
catalyst; Irisin, human recombinant; Runx2, Runt-related
transcription factor 2; PI3K, phosphatidylinositol-3-kinase; Akt,
protein kinase B; mTOR, mammalian target of rapamycin; PGC-1α,
peroxisome proliferators-activated receptor γ coactivator 1α;
RANKL, receptor activator of nuclear factor-κB ligand; NFATc1,
nuclear factor of activated T cells; MAPK, mitogen-activated
protein kinase; ERK, extracellular signal-regulated kinase; BMP-2,
bone morphogenetic protein type 2; Smad3, mothers against
decapentaplegic homolog 3; Wnt, wingless-related integration
site.

Figure 2

Regulatory pathways of muscle factors on the musculoskeletal system. IGF-1 modulates muscle metabolism via the PI3K/Akt/mTOR pathway and bone metabolism through PI3K/Akt and MAPK/ERK signaling. FGF-2 regulates muscle metabolism via MAPK/ERK and bone metabolism through BMP-2/Smad signaling. Myostatin influences muscle metabolism via Smad3 and P21, and bone metabolism through Smad2 and Wnt/β-catenin pathways. PGC-1α stimulates PNDC5, promoting Irisin release from muscle; Irisin subsequently regulates bone metabolism via P38MAPK/ERK and RANKL/NFATc1 signaling. IGF-1, Insulin-like growth factor-1; FGF-2, Fibroblast growth factor 2; GSK3, Glycogen Synthase Kinase-3; IGFBP-3, insulin-like growth factor binding protein 3; IGFBP-5, insulin-like growth factor binding protein 5; MyoD, Myoblast determination protein 1; PNDC, peroxynitrite decomposition catalyst; Irisin, human recombinant; Runx2, Runt-related transcription factor 2; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; PGC-1α, peroxisome proliferators-activated receptor γ coactivator 1α; RANKL, receptor activator of nuclear factor-κB ligand; NFATc1, nuclear factor of activated T cells; MAPK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; BMP-2, bone morphogenetic protein type 2; Smad3, mothers against decapentaplegic homolog 3; Wnt, wingless-related integration site.

Regulatory mechanism of estrogen on
the musculoskeletal system. Estrogen modulates bone metabolism
through PI3K/Akt, MAPK/ERK, RANKL/OPG, and Wnt/β-catenin pathways.
In muscle, estrogen influences metabolism via PI3K/Akt/mTOR,
GPER/MAPK/ERK, RANKL/OPG, and MuRF1/MAFbx pathways. GPER, G
protein-coupled estrogen receptor; ERβ, estrogen receptor beta;
ERα, estrogen receptor alpha; Runx2, Runt-related transcription
factor 2; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase
B; mTOR, mammalian target of rapamycin; MAPK, mitogen-activated
protein kinase; ERK, extracellular signal-regulated kinase; RANKL,
receptor activator of nuclear factor-κB ligand; OPG,
osteoprotegerin; MAFbx, muscle atrophy F-box; MuRF1, muscle RING
finger-1.

Figure 3

Regulatory mechanism of estrogen on the musculoskeletal system. Estrogen modulates bone metabolism through PI3K/Akt, MAPK/ERK, RANKL/OPG, and Wnt/β-catenin pathways. In muscle, estrogen influences metabolism via PI3K/Akt/mTOR, GPER/MAPK/ERK, RANKL/OPG, and MuRF1/MAFbx pathways. GPER, G protein-coupled estrogen receptor; ERβ, estrogen receptor beta; ERα, estrogen receptor alpha; Runx2, Runt-related transcription factor 2; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; MAPK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; RANKL, receptor activator of nuclear factor-κB ligand; OPG, osteoprotegerin; MAFbx, muscle atrophy F-box; MuRF1, muscle RING finger-1.

Molecular signaling pathways in musculoskeletal crosstalk mediated by IGF-1 and FGF-2
Mechanisms of IGF-1-mediated signaling pathways in musculoskeletal crosstalk

As a pivotal growth factor in muscle-bone communication, IGF-1 regulates musculoskeletal crosstalk through multiple signaling pathways (Fig. 4) (38). Upon binding to its receptor, IGF-1 activates the PI3K/Akt pathway, promoting muscle fiber proliferation and repair while modulating bone cell metabolism (39). In the skeletal system, the PI3K/Akt pathway influences osteoblast and osteoclast activity via downstream mTOR signaling, thus regulating bone formation and resorption (40). Activation of PI3K/Akt leads to Akt kinase phosphorylation, which in turn activates proteins involved in cell growth and metabolism, promoting proliferation, survival and differentiation (41). In bone, the PI3K/Akt pathway is essential for bone formation by stimulating osteoblast proliferation and differentiation, while enhancing bone matrix mineralization (42). IGF-1 activation of this pathway improves bone density and accelerates fracture healing, highlighting its role in bone repair (43).

Regulatory mechanism of IGF-1 on the
musculoskeletal system. IGF-1 modulates bone metabolism through
several signaling pathways, including Wnt/β-catenin, PI3K/Akt,
MAPK, RANKL/OPG, and Akt. It also influences muscle metabolism via
the PI3K/Akt/mTOR, MuRF1/MAFbx, and GLUT4 pathways. Wnt,
ςingless-type MMTV integration site family; Bcl-2, B-cell
lymphoma-2; GH, growth hormone; IGF-1, insulin-like growth factor
1; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B;
mTOR, mammalian target of rapamycin; MAPK, mitogen-activated
protein kinase; ERK, extracellular signal-regulated kinase; RANKL,
receptor activator of nuclear factor-κB ligand; OPG,
osteoprotegerin; MAFbx, muscle atrophy F-box; MuRF1, muscle RING
finger-1; GLUT4, Glucose transporter protein 4.

Figure 4

Regulatory mechanism of IGF-1 on the musculoskeletal system. IGF-1 modulates bone metabolism through several signaling pathways, including Wnt/β-catenin, PI3K/Akt, MAPK, RANKL/OPG, and Akt. It also influences muscle metabolism via the PI3K/Akt/mTOR, MuRF1/MAFbx, and GLUT4 pathways. Wnt, ςingless-type MMTV integration site family; Bcl-2, B-cell lymphoma-2; GH, growth hormone; IGF-1, insulin-like growth factor 1; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; MAPK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase; RANKL, receptor activator of nuclear factor-κB ligand; OPG, osteoprotegerin; MAFbx, muscle atrophy F-box; MuRF1, muscle RING finger-1; GLUT4, Glucose transporter protein 4.

The role of signal transduction pathways involved in FGF-2 [such as FGF receptor (FGFR)-related pathways] in musculoskeletal interactions

FGF-2 contributes to musculoskeletal crosstalk by promoting muscle growth and regulating bone formation and repair through the FGFR (Fig. 5) (44,45). In bone metabolism, FGF-2 modulates osteoblast and osteoclast functions, influencing bone formation and resorption via the MAPK/ERK signaling pathway (46). Research shows that FGF-2 enhances bone repair by stimulating osteoblast proliferation and migration, accelerating new bone formation during fracture healing (47). Additionally, FGF-2 helps maintain bone density by balancing osteoblast and osteoclast activity, thus reducing bone resorption (48). FGF-2 exerts a bidirectional regulatory effect in musculoskeletal crosstalk, promoting bone formation while inhibiting excessive bone resorption. In summary, both IGF-1 and FGF-2 are crucial in musculoskeletal interactions, regulating bone and muscle growth and repair through their respective signaling pathways and coordinating the development and repair of the musculoskeletal system through their interplay.

Regulatory mechanism of FGF-2 on the
musculoskeletal system. FGF-2 regulates bone metabolism through the
ERK, JAK-STAT, PI3K/Akt, and RANKL/OPG pathways. In muscle, it
affects metabolism via AMPK/PGC-1α, AMPK/SIRT-1, PGC-1α/GLUT4,
PGC-1α/IRS-1, FOXO-3/Atrogin-1 and FOXO-3/MuRF-1 signaling. FGF-2,
Basic fibroblast growth factor; JAK, Janus kinase; STAT, Signal
transducers and activators of transcription; PI3K,
phosphatidylinositol-3-kinase; Akt, protein kinase B; mTOR,
mammalian target of rapamycin; Atrogin-1, muscle atrophy-1; PR,
progesterone receptors; FGFR, fibroblast growth factor receptor;
AMPK, AMP-activated protein kinase; PGC-1α, peroxisome
proliferator-activated receptor gamma coactivator 1 alpha; MAPK,
Mitogen-activated protein kinase; ERK, Extracellular
signal-regulated kinase; RANKL, receptor activator of nuclear
factor-κB ligand; OPG, Osteoprotegerin; MuRF1, muscle RING
finger-1; GLUT4, Glucose transporter protein 4; SIRT1, Sirtuin1;
IRS1, Insulin receptor substrate 1; FOXO-3, Forkhead box O3.

Figure 5

Regulatory mechanism of FGF-2 on the musculoskeletal system. FGF-2 regulates bone metabolism through the ERK, JAK-STAT, PI3K/Akt, and RANKL/OPG pathways. In muscle, it affects metabolism via AMPK/PGC-1α, AMPK/SIRT-1, PGC-1α/GLUT4, PGC-1α/IRS-1, FOXO-3/Atrogin-1 and FOXO-3/MuRF-1 signaling. FGF-2, Basic fibroblast growth factor; JAK, Janus kinase; STAT, Signal transducers and activators of transcription; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin; Atrogin-1, muscle atrophy-1; PR, progesterone receptors; FGFR, fibroblast growth factor receptor; AMPK, AMP-activated protein kinase; PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha; MAPK, Mitogen-activated protein kinase; ERK, Extracellular signal-regulated kinase; RANKL, receptor activator of nuclear factor-κB ligand; OPG, Osteoprotegerin; MuRF1, muscle RING finger-1; GLUT4, Glucose transporter protein 4; SIRT1, Sirtuin1; IRS1, Insulin receptor substrate 1; FOXO-3, Forkhead box O3.

Macrophage-derived exosomes

Exosomes are small, bilayer lipid-enclosed vesicles secreted by cells, encompassing exosomes, microvesicles and apoptotic bodies (7). Among these, exosomes are the most extensively studied subtype (49). They carry a variety of cargo, including proteins, metabolites, nucleic acids and lipids, derived from their parent cells, exerting biological effects similar to those of the original cells (Table IV) (50-53). Exosomes can bind to recipient cells through surface ligands or be internalized via paracrine signaling and membrane fusion (such as endocytosis) (54), facilitating the transfer of bioactive molecules and modulating recipient cell functions. Macrophage-derived exosomes play critical roles in regulating inflammation and influencing the progression of bone diseases, such as osteoporosis, fractures and osteoarthritis, markedly affecting bone metabolism and homeostasis (55-57). Research indicates that both the composition and quantity of exosomes vary depending on the macrophage polarization state (58-60). In recent years, macrophage-derived exosomes have gained growing research interest, following extensive earlier studies on exosomes from mesenchymal stem cells (MSCs).

Table IV

Comprehensive composition and functions of macrophage-derived exosomal components.

Table IV

Comprehensive composition and functions of macrophage-derived exosomal components.

Component categoryRepresentative moleculesBiological functions
Proteins• MHC class II (HLA-DR)
• Integrins (αMβ2)
• Heat shock proteins (HSP70/90)
• Antigen presentation
• Tissue-specific homing
• Protein folding and stress response
Nucleic acids• miRNAs (such as miR-155, miR-21)
• lncRNAs (such as H19)
• mtDNA
• Post-transcriptional gene regulation
• Epigenetic modulation
• cGAS-STING pathway activation
Lipids• Sphingomyelin (SM)
• Cholesterol
• Prostaglandin E2 (PGE2)
• Membrane stability and curvature
• Inflammatory signaling modulation
• Vascular permeability regulation
Metabolites• Lactate
• Succinate
• Glutathione (GSH)
• Glycolytic metabolic signaling
• Pro-inflammatory signal amplification
• Redox homeostasis maintenance
Receptors/signalig• TLR4
• TNF-α
• TGF-β1
• Pathogen-associated molecular pattern recognition
• Pro-/anti-inflammatory signal transduction
• Immunomodulation and tissue repair
(Refs.)(50-53)

[i] MHC, major histocompatibility complex; HLA-DR, Human histocompatibility leukocyte antigen (HLA)-DR; cGAS, Cyclic GMP-AMP synthase; STING, Stimulator of interferon genes; TGF-β1, Transforming growth factor-beta 1; TNF-α, Tumor necrosis factor alpha; TLR4, Toll-like receptor 4; miRNAs, microRNAs; lncRNAs, long noncoding RNAs; mtDNA, Mitochondrial genome.

Macrophage-derived exosomes and their effect on musculoskeletal metabolism
The role of M1 macrophage-derived exosomes in bone metabolism

M1-polarized macrophages promote the release of chemotactic and inflammatory mediators, which facilitate osteoclast activation and debris clearance at fracture sites (61-63). MicroRNAs (miRNAs), highly conserved non-coding RNAs, post-transcriptionally regulate gene expression and play a key role in intercellular communication as integral components of exosomes (64). The miRNA profiles of M1 and M2 polarized macrophages exhibit significant differences. Kang et al (59) demonstrated that M1 exosomes are enriched with miR-155, which inhibits the bone morphogenetic protein (BMP) signaling pathway by downregulating BMP2, BMP9, and Runt-related transcription factor 2 (Runx2). Given that BMP2 is critical for osteoblast differentiation (65), its downregulation impedes bone regeneration. Ge et al (60) reported that exosomal miR-155 from M1 macrophages targets SOCS6, resulting in elevated p65 protein levels. This activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway exacerbates inflammation. Additionally, miR-222 is highly expressed in M1 macrophages (66). Studies have shown that exosomal miR-222 from M1 macrophages induces apoptosis in bone marrow mesenchymal stem cells (BMMSCs) by inhibiting Bcl-2 expression (67-69). Yu et al (70) demonstrated that exosomal miR-98 from M1 macrophages targets DUSP1 in MC3T3-E1 cells and a postmenopausal osteoporosis murine model, inhibiting osteogenic differentiation. Another study showed that M1 exosomes are enriched with miR-1246, which activates the Wnt pathway by downregulating GSK3β and Axin2, thereby promoting cartilage inflammation and degradation (71). These findings were primarily validated using bioinformatics analyses, sequencing and related methodologies. In summary, M1 macrophage-derived exosomes, which are enriched with miRNAs such as miR-155, miR-222, miR-98 and miR-1246, modulate downstream signaling pathways to promote inflammation and impair bone repair both in vitro and in vivo. The specific miRNA cargo determines the downstream signaling effects by targeting key nodes within signaling networks. Exosomes serve as specific carriers, enabling macrophage-derived miRNAs to function as promising biomarkers for monitoring and regulating bone remodeling. However, the complete repertoire of miRNAs and other cargo within macrophage exosomes remains incompletely characterized. Further investigation is needed to understand how macrophage polarization influences exosomal miRNA cargo and the precise mechanisms through which these miRNAs regulate downstream pathways. The effects of macrophage exosomes on BMMSC differentiation have been extensively studied. He et al (72) collected conditioned medium (CM) from M1 macrophages and observed enhanced BMMSC proliferation, adipogenic differentiation and extracellular matrix deposition. Upon isolating exosomes from this CM, it was found that M1 exosomes, but not M2 exosomes, promoted stem cell proliferation, osteogenesis and adipogenesis. This finding was corroborated by Xia et al (73). By contrast, Kang et al (59) reported that co-culture with M1 macrophages markedly reduced BMP2 and BMP9 expression in mouse MSCs, suggesting that M1 macrophage-derived exosomes impair the osteoinductive effects of BMPs and suppress osteogenic gene expression in MSCs. Through exosomal communication, M1 macrophages primarily enhance early and mid-stage osteogenesis, exerting stronger effects on BMMSC proliferation, osteogenesis and adipogenesis compared with M2 macrophages. These differing effects on osteogenesis may be attributed to variations in recipient cell sources. Experimental discrepancies could stem from multiple factors, including macrophage maturity, co-culture conditions, the presence of other cytokines in the CM, recipient cell lineage, and technical differences in exosome isolation from specific conditioned media (62,73). The balance between osteogenic and adipogenic differentiation in BMMSCs is essential for maintaining bone mass. Investigating the mechanisms by which M1 macrophage-derived exosomes regulate this balance in BMMSCs may offer insights into clinical disease pathogenesis and aid in the development of targeted therapies. Fig. 6 summarizes the regulation of bone remodeling signaling pathways by M1 macrophage-derived exosomes.

Regulation of bone remodeling
signaling pathways by M1 exosomes. M1 macrophage-derived exosomes
contain specific microRNAs, including miR-1246, miR-98, miR-122 and
miR-155, that regulate bone metabolism by targeting key signaling
molecules. Specifically, miR-1246 downregulates GSK3B and Axin2;
miR-98 suppresses DUSP-1; miR-122 inhibits Bcl-2 and miR-155
represses both BMP and SOCS6. Additionally, these exosomes modulate
bone remodeling through the NF-κB signaling pathway. miRNAs/miRs,
microRNAs; M1 Exo, M1 macrophage-derived exosomes; Omap, Office of
Medical Assistance Programs; GSK3B, Glycogen synthase kinase 3
beta; DUSP-1, Dual specificity phosphatase 1; Bcl-2, B-cell
lymphoma/Leukemia-2; BMP, Bone morphogenetic protein; Axin2, Axis
inhibition protein 2; JNK, c-Jun N-terminal kinase; SOCS6,
suppressor of cytokine signaling 6; BMMSC, Bone marrow mesenchymal
stem cells; NF-κB, Nuclear factor kappa B.

Figure 6

Regulation of bone remodeling signaling pathways by M1 exosomes. M1 macrophage-derived exosomes contain specific microRNAs, including miR-1246, miR-98, miR-122 and miR-155, that regulate bone metabolism by targeting key signaling molecules. Specifically, miR-1246 downregulates GSK3B and Axin2; miR-98 suppresses DUSP-1; miR-122 inhibits Bcl-2 and miR-155 represses both BMP and SOCS6. Additionally, these exosomes modulate bone remodeling through the NF-κB signaling pathway. miRNAs/miRs, microRNAs; M1 Exo, M1 macrophage-derived exosomes; Omap, Office of Medical Assistance Programs; GSK3B, Glycogen synthase kinase 3 beta; DUSP-1, Dual specificity phosphatase 1; Bcl-2, B-cell lymphoma/Leukemia-2; BMP, Bone morphogenetic protein; Axin2, Axis inhibition protein 2; JNK, c-Jun N-terminal kinase; SOCS6, suppressor of cytokine signaling 6; BMMSC, Bone marrow mesenchymal stem cells; NF-κB, Nuclear factor kappa B.

The role of M2 macrophage-derived exosomes in bone metabolism

A critical event in bone healing is the transition from the pro-inflammatory M1 macrophage phenotype to the anti-inflammatory M2 phenotype (74). In the later stages of bone healing, M2 macrophages establish an anti-inflammatory environment that promotes osteogenesis in BMMSCs. Li et al (56) identified exosomes as key mediators of the osteogenic process induced by M2 macrophages. BMPs are key endogenous regulators of osteogenesis, with the loss of BMP2 or BMP4 function leading to significant impairments in osteogenesis. BMPs activate Smad proteins and upregulate Runx2 expression, driving MSC differentiation into osteoblasts (75,76). Using an in vivo bone defect model, Kang et al (59) demonstrated that M2 macrophage-derived exosomes promote bone regeneration. Further analysis revealed that M2 exosomes are enriched with miR-378a, which enhances osteoinduction by modulating the BMP signaling pathway, thus promoting cranial bone regeneration in mice. Studies also indicate that exosomes from IL-4-stimulated M2 macrophages, enriched with miRNAs such as miR-99a-5p, miR-146b-5p and miR-378-3p, suppress inflammation by downregulating the TNF-α and NF-κB signaling pathways in Apoe−/− mice (77). Moreover, exosomes derived from M2 macrophages promote osteogenic differentiation of BMMSCs through miR-690, IRS-1 and TAZ, while inhibiting adipogenic differentiation (56). Yu et al (78) reported that miR-690, enriched in M2 macrophage-derived exosomes, upregulates osteogenic differentiation in C2C12 myogenic progenitor cells by inhibiting the translation of the NF-κB p65 protein. M2 macrophage-derived exosomes are also shown to carry high levels of miR-221-5p, which promotes tissue repair and regeneration by binding to the 3' untranslated region (UTR) of E2F2 mRNA and negatively regulating its expression in pancreatic ductal adenocarcinoma (79). Collectively, these studies suggest that M2 macrophage-derived exosomes infiltrate the bone marrow microenvironment and transfer specific miRNAs (such as miR-378a, miR-690, miR-99a-5p) to BMMSCs, facilitating osteogenic differentiation and fracture healing (80,81). These miRNAs likely function as pro-osteogenic agents by modulating key pathways such as BMP signaling to enhance osteogenesis. However, the precise roles of M2 exosome-derived miRNAs in bone formation and osteoclastogenesis remain to be fully elucidated (82). Emerging evidence suggests that interactions between BMMSCs and extracellular signals from M2 macrophages markedly contribute to bone formation. However, the exact mechanisms underlying these interactions remain debated, possibly due to differences in cell sources, co-culture conditions, and macrophage polarization protocols. Some studies propose that CM from M0 or M2 macrophages promotes the mineralization of BMMSCs in the later stages of osteogenesis (83-86). By contrast, other studies report opposing findings. For instance, Xia et al (73) found that exosomes derived from M2 macrophages inhibit BMMSC proliferation, with no significant effect on the expression of osteogenic or adipogenic genes in BMMSCs, and possibly even suppress chondrogenic differentiation. The regulatory effects of M2 macrophage-derived exosomes on bone remodeling signaling pathways are summarized in Fig. 7.

Regulation of bone remodeling
signaling pathways by M2-derived exosomes. Exosomes derived from M2
macrophages carry miRNAs such as miR-378a, miR-99a, miR-146b,
miR-378a, and miR-690, which positively influence bone metabolism.
These miRNAs act through multiple targets, miR-378a enhances BMP
signaling; the miR-99a/146b/378a cluster inhibits NF-κB; and
miR-690 suppresses both NF-κB p65 and IRS-1 while promoting TAZ
activity. Furthermore, M2 exosomes contribute to bone regulation by
modulating the behavior of BMMSCs. miRNAs/miRs, microRNAs; BMP,
Bone morphogenetic protein(s); NF-κB, Nuclear factor kappa B; IRS1,
Insulin receptor substrate 1; OCN, osteocalcin; TAZ,
Transcriptional co-activator with PDZ-binding motif; BMMSCs, Bone
marrow mesenchymal stem cells.

Figure 7

Regulation of bone remodeling signaling pathways by M2-derived exosomes. Exosomes derived from M2 macrophages carry miRNAs such as miR-378a, miR-99a, miR-146b, miR-378a, and miR-690, which positively influence bone metabolism. These miRNAs act through multiple targets, miR-378a enhances BMP signaling; the miR-99a/146b/378a cluster inhibits NF-κB; and miR-690 suppresses both NF-κB p65 and IRS-1 while promoting TAZ activity. Furthermore, M2 exosomes contribute to bone regulation by modulating the behavior of BMMSCs. miRNAs/miRs, microRNAs; BMP, Bone morphogenetic protein(s); NF-κB, Nuclear factor kappa B; IRS1, Insulin receptor substrate 1; OCN, osteocalcin; TAZ, Transcriptional co-activator with PDZ-binding motif; BMMSCs, Bone marrow mesenchymal stem cells.

Effects of macrophage-derived exosomes on muscle growth and metabolism

Exosomes play a pivotal role in regulating protein synthesis and muscle regeneration by promoting satellite cell proliferation and myofiber formation. This regulation occurs through the delivery of myogenic growth factors such as IGF, FGF-2, and hepatocyte growth factor (HGF) (87). Forterre et al (88) employed proteomic analysis to identify numerous proteins associated with muscle growth and metabolism in exosomes secreted by skeletal muscle. For example, exosomes extracted from C2C12-derived myotubes contained key components, such as integrin subunit β1, CD9, CD81, neural cell adhesion molecule (NCAM), CD44 and myosin, that are potentially crucial for myocyte differentiation. Similarly, Mobley et al (89) demonstrated that exosomes derived from whey protein enhance muscle protein synthesis in vitro. Autophagy plays a critical role in maintaining muscle mass and function; its inhibition exacerbates muscle atrophy (90). AMPK, a central intracellular energy sensor, alleviates sarcopenia (SP) symptoms and mitochondrial dysfunction (91). Chen et al (92) discovered that exosomes from MSCs ameliorate muscle atrophy by enhancing AMPK/ULK1-mediated autophagy. Guescini et al (93) observed that exosomal miR-133b and miR-181a-5p are rapidly released into the bloodstream post-exercise, promoting muscle regeneration. This mechanism may partially explain how exercise mitigates SP. Furthermore, Chaturvedi et al (94) demonstrated in animal models that exercise-induced exosomes promote skeletal muscle regeneration through the upregulation of miR-9b and miR-29.

Additionally, extracellular vesicles from M1 macrophages can polarize recipient macrophages toward an M2-like phenotype, thereby altering skeletal muscle homeostasis under high-glucose conditions. Collectively, these findings highlight the roles of both M1- and M2-derived macrophage exosomes in regulating SP.

The role of exosomes from macrophages in muscle-bone crosstalk

Exosomes serve as critical messengers in muscle-bone crosstalk, enabling skeletal muscles to influence bone activity through EV-mediated communication. This exosome-mediated communication between skeletal muscle cells occurs via autocrine, paracrine, or endocrine pathways (95-98). Exosomes derived from C2C12 myoblasts promote the osteogenic differentiation of MC3T3-E1 preosteoblasts, likely mediated by the upregulation of miR-27a-3p in recipient cells. This miRNA suppresses specific target genes and activates β-catenin signaling, thereby enhancing osteogenesis (99). Additionally, exosomes from C2C12 myoblasts enhance Wnt/β-catenin signaling in TOPflash-MLOY4 osteocyte-like cells, promoting cell survival and providing protection against apoptosis and oxidative stress. This protective effect is likely due to exosomes-mediated inactivation of Wnt inhibitors, such as SOST, DKK2, and SFRP2 (100). Li et al (101) showed that myoblast-derived exosomes carry Prrx2, which activates miR22HG transcription, promoting osteogenic differentiation of BMMSCs via the YAP pathway and miR-128. Furthermore, CXCR4, present on exosomes produced by mouse fibroblasts, targets these vesicles to the bone marrow, where miR-188-containing exosomes fuse with lipids to form hybrid nanoparticles. These nanoparticles then release miR-188 in a targeted manner, inhibiting adipogenesis and promoting BMMSC differentiation into osteoblasts (102). Regarding the influence of bone cell-derived exosomes on skeletal muscle, long non-coding RNAs (lncRNAs) are critical. For example, Zheng et al (103) found that osteoblasts induce myogenic differentiation of C2C12 myoblasts via exosomal lncRNAs, specifically TUG1 and DANCR. Toita et al (104) demonstrated that collagen patches releasing phosphatidylserine-containing liposomes promote M1-to-M2 macrophage polarization, facilitating concurrent bone and muscle tissue healing. Transcriptome analysis via next-generation sequencing revealed that M1 macrophage secretory products inhibit the differentiation of preosteoblasts and myoblasts, while M2 macrophage secretory products promote it. This highlights the importance of timely M1-to-M2 polarization for effective tissue regeneration. As shown in Fig. 8, macrophages, particularly the M2 phenotype, release exosomes carrying the transcription factor C/EBPβ. This factor promotes skeletal muscle cell proliferation and differentiation via the PPARγ signaling pathway (105). Depletion of macrophage exosomes containing C/EBPβ markedly inhibits ALP activity in muscle cells and reduces the expression of myogenic markers (such as MyoD and MyoG), delaying muscle injury repair (105). M2 macrophage-derived exosomes suppress M1 macrophage activity by delivering anti-inflammatory factors, such as IL-10 and TGF-β, while mitigating the damaging effects of pro-inflammatory factors like TNF-α and IL-6 on muscle fibers. They also promote the directed differentiation of stem cells into myocytes (106). Additionally, M2 macrophage exosomes activate the PI3K/AKT pathway, facilitating the shift from M1 to M2 polarization. This process leads to reduced inflammatory cytokine levels, improved bone immune microenvironment and enhanced osteoblast differentiation and angiogenesis (107). Under mechanical stimulation, macrophages increase exosome secretion. These exosomes carry the UCHL3 protein, which enhances the osteogenic potential of BMMSCs and drives callus formation through the SMAD1 signaling pathway (108). Exosome-encapsulated miRNAs, including miR-21 and miR-148a, inhibit osteogenic inhibitors like PTEN and upregulate RUNX2 and osterix (106). In osteosarcoma models, exosomes coordinate osteoclast and osteoblast activity by transmitting Wnt/β-catenin signals, contributing to the maintenance of bone homeostasis (106). These findings highlight the critical role of macrophage-derived exosomes in regulating the proliferation and repair of both bone and muscle tissues, modulating muscle-bone crosstalk and enhancing tissue regeneration.

Mechanism of macrophage-derived
exosomes on bone metabolism and musculoskeletal crosstalk. M2
macrophage-derived exosomes regulate muscle metabolism through
several pathways, the miR-23a/MuRF1/MAFbx axis, C/EBP-β/PPARγ
signaling, miR-206/HDAC4, Akt/mTOR signaling, and by modulating M1
macrophage activity. Concurrently, macrophage-derived exosomes
regulate bone metabolism via the PI3K/Akt and Wnt/β-catenin
pathways, miR-1481/PTEN and miR-21/PTEN axes, and the UCHL3/SMAD1
signaling cascade. miRNAs/miRs, microRNAs; PPARγ, Peroxisome
proliferator-activated receptor gamma; mTOR, mammalian target of
rapamycin; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase
B; MuRF1, muscle RING finger-1; MAFbx, F-box only protein 32;
Smad1, SMAD family member 1; Wnt, wingless-type; MMTV integration
site family; TGF-β, transforming growth factor beta; HDAC4, histone
deacetylation 4; C/EBP-β, CCAAT-enhancer binding protein; UCHL3,
ubiquitin carboxyl terminal hydrolase L3; ALP, alanine phosphatase;
MyoD, myogenic differentiation 1; MYOG, Human myogenin; BMMSC, Bone
marrow stromal cells; PTEN, Phosphatase and tensin homolog; RUNX2,
Runt-related transcription factor 2.

Figure 8

Mechanism of macrophage-derived exosomes on bone metabolism and musculoskeletal crosstalk. M2 macrophage-derived exosomes regulate muscle metabolism through several pathways, the miR-23a/MuRF1/MAFbx axis, C/EBP-β/PPARγ signaling, miR-206/HDAC4, Akt/mTOR signaling, and by modulating M1 macrophage activity. Concurrently, macrophage-derived exosomes regulate bone metabolism via the PI3K/Akt and Wnt/β-catenin pathways, miR-1481/PTEN and miR-21/PTEN axes, and the UCHL3/SMAD1 signaling cascade. miRNAs/miRs, microRNAs; PPARγ, Peroxisome proliferator-activated receptor gamma; mTOR, mammalian target of rapamycin; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; MuRF1, muscle RING finger-1; MAFbx, F-box only protein 32; Smad1, SMAD family member 1; Wnt, wingless-type; MMTV integration site family; TGF-β, transforming growth factor beta; HDAC4, histone deacetylation 4; C/EBP-β, CCAAT-enhancer binding protein; UCHL3, ubiquitin carboxyl terminal hydrolase L3; ALP, alanine phosphatase; MyoD, myogenic differentiation 1; MYOG, Human myogenin; BMMSC, Bone marrow stromal cells; PTEN, Phosphatase and tensin homolog; RUNX2, Runt-related transcription factor 2.

Mechanism of influence of macrophage-derived exosomes on bone metabolism of musculoskeletal crosstalk mediated by IGF-1 and FGF-2

Key molecules in macrophage-derived exosomes
Regulation of IGF-1 and FGF-2 signaling by exosomal miRNA and protein components

Exosomes function as essential intercellular signaling vehicles, transporting a variety of biologically active molecules, including miRNAs, mRNAs, proteins and lipids (109). These components regulate bone metabolism by modulating signaling pathways in target cells (110). Notably, miRNAs within macrophage-derived exosomes are recognized as key regulatory factors (111). Research indicates that macrophage-derived exosomal miRNAs influence bone metabolism indirectly by regulating key signaling molecules such as IGF-1 and FGF-2 (Figs. 9 and 10) (112). miRNAs modulate gene expression by binding to target mRNAs, resulting in translational repression or mRNA degradation (113). In musculoskeletal crosstalk, exosomal miRNAs from macrophages play pivotal roles in the regulation of IGF-1, FGF-2 and other signaling pathways (Figs. 9 and 10) (114). For instance, macrophage-derived exosomal miR-21 promotes IGF-1 signaling by targeting inhibitory molecules within the PI3K-Akt pathway, thereby enhancing bone formation (115). Similarly, macrophage-derived exosomal miR-29 boosts osteoblast function, promoting bone matrix synthesis and mineralization through the regulation of FGF-2 expression (116). By contrast, M1 exosomal miR-155 directly inhibits IGF-1R and downstream PI3K/Akt signaling, diminishing osteoblast differentiation (117). Additionally, M1 exosomal miR-143 exacerbates insulin-resistant muscle atrophy by inhibiting IRS-1 and obstructing the pro-muscle protein synthesis effect of IGF-1 (118). Thus, macrophage-derived exosomes regulate musculoskeletal crosstalk via exosomal miRNAs during IGF-1 and FGF-2 signaling, revealing a novel molecular mechanism (Figs. 9 and 10).

Mechanism of macrophage-derived
exosomes on bone metabolism and musculoskeletal crosstalk by
regulating IGF-1 expression. M2 macrophage-derived exosomes are
enriched with miR-29a, circ-Eif3c, and IGF-1. miR-29a and
circ-Eif3c regulate bone and muscle metabolism by modulating the
IGF-1/PI3K/Akt signaling pathway. Independently, IGF-1 regulates
muscle metabolism by inhibiting the MuRF1/MAFbx and NF-κB pathways.
IGF-1, insulin-like growth factor 1; miRNAs/miRs, microRNAs; circ,
circular RNA; PI3K, phosphatidylinositol-3-kinase; Akt, protein
kinase B; MuRF1, muscle RING finger-1; MAFbx, F-box only protein
32; NF-κB, Nuclear factor kappa B.

Figure 9

Mechanism of macrophage-derived exosomes on bone metabolism and musculoskeletal crosstalk by regulating IGF-1 expression. M2 macrophage-derived exosomes are enriched with miR-29a, circ-Eif3c, and IGF-1. miR-29a and circ-Eif3c regulate bone and muscle metabolism by modulating the IGF-1/PI3K/Akt signaling pathway. Independently, IGF-1 regulates muscle metabolism by inhibiting the MuRF1/MAFbx and NF-κB pathways. IGF-1, insulin-like growth factor 1; miRNAs/miRs, microRNAs; circ, circular RNA; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; MuRF1, muscle RING finger-1; MAFbx, F-box only protein 32; NF-κB, Nuclear factor kappa B.

Mechanism of macrophage-derived
exosomes on bone metabolism and musculoskeletal crosstalk by
regulating FGF-2 expression. Macrophage exosomes contain miR-21-5p,
miR-29a, lncRNA H19, and FGF-2. The miRNAs and lncRNA H19 regulate
muscle metabolism by targeting FGF-2, activating PI3K/Akt
signaling, and promoting the expression of myogenic factors MyoD
and Myogenin. FGF-2 regulates both muscle and bone metabolism
through the TGF-β/SMAD3, PI3K/Akt/mTOR, and FOXO-3 pathways.
miRNAs/miRs, microRNAs; lnc, long non-coding; FGF-2, basic
fibroblast growth factor; PI3K, phosphatidylinositol-3-kinase; Akt,
protein kinase B; MuRF1, muscle RING finger-1; MyoD, Myogenic
differentiation 1; ERK, extracellular regulated protein kinases;
RUNX2, Runt-related transcription factor 2; TGF-β, transforming
growth factor beta; SMAD3, Mothers against decapentaplegic homolog
3; mTOR, mammalian target of rapamycin; FOXO, forkhead box O;
Atrogin-1, muscle atrophy-1.

Figure 10

Mechanism of macrophage-derived exosomes on bone metabolism and musculoskeletal crosstalk by regulating FGF-2 expression. Macrophage exosomes contain miR-21-5p, miR-29a, lncRNA H19, and FGF-2. The miRNAs and lncRNA H19 regulate muscle metabolism by targeting FGF-2, activating PI3K/Akt signaling, and promoting the expression of myogenic factors MyoD and Myogenin. FGF-2 regulates both muscle and bone metabolism through the TGF-β/SMAD3, PI3K/Akt/mTOR, and FOXO-3 pathways. miRNAs/miRs, microRNAs; lnc, long non-coding; FGF-2, basic fibroblast growth factor; PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; MuRF1, muscle RING finger-1; MyoD, Myogenic differentiation 1; ERK, extracellular regulated protein kinases; RUNX2, Runt-related transcription factor 2; TGF-β, transforming growth factor beta; SMAD3, Mothers against decapentaplegic homolog 3; mTOR, mammalian target of rapamycin; FOXO, forkhead box O; Atrogin-1, muscle atrophy-1.

Interaction between exosome components and musculoskeletal cell surface receptors

Exosomes carry a diverse array of cargo molecules that regulate bone and muscle metabolism through interactions with receptors on the surface of musculoskeletal cells (119). Signal transduction by IGF-1 and FGF-2 in bone metabolism is initiated when these factors bind to their specific cell surface receptors (120). Macrophage-derived exosomes can modulate bone metabolism by transporting receptors or receptor ligands that facilitate the binding of signaling factors to their target cells (121). Exosomal ligands for IGF-1R and FGFR directly bind to their corresponding receptors on bone or muscle cells, triggering signal transduction (122). Upon IGF-1 binding, IGF-1R activates intracellular signaling cascades, primarily via the PI3K/Akt and MAPK pathways, to regulate bone and muscle growth and repair (123,124). Macrophage-derived exosomes can deliver IGF-1R or its ligands, thereby enhancing IGF-1 signaling to promote bone formation and muscle repair (Fig. 9) (125). Similarly, FGF-2 binding to FGFR initiates signaling cascades that regulate the proliferation and differentiation of bone marrow stromal cells and osteoblasts (126). FGF-2 targets sclerostin in bone and myostatin in skeletal muscle to counteract the harmful effects of glucocorticoids on musculoskeletal degradation (127). The exosome-mediated interaction between FGF-2 and FGFR enhances bone repair and plays a pivotal role in musculoskeletal crosstalk (Fig. 10) (128). In summary, macrophage-derived exosomes facilitate signal transduction and regulate bone metabolism by transporting receptors (such as IGF-1R, FGFR) and their ligands, strengthening bone formation and repair, and promoting musculoskeletal crosstalk.

The effect on osteoblast function
Regulating the proliferation, differentiation, and mineralization of osteoblasts

Osteoblast proliferation, differentiation and mineralization are essential processes in bone metabolism, regulated by macrophage-derived exosomes through various mechanisms (129). During osteogenesis, active molecules such as miRNAs and proteins facilitate osteoblast proliferation, differentiation, and mineralization (130). Exosomal miRNAs secreted by macrophages, including miR-21 and miR-29, can activate the PI3K/Akt and Wnt/β-catenin signaling pathways in osteoblasts, promoting their proliferation and differentiation (131); M1 macrophage-derived exosomes aggravate bone loss in postmenopausal osteoporosis via a miR-98/dual specificity phosphatase 1 (Dusp1)/c-Jun N-terminal kinase (JNK) axis (132); M2 macrophagy-derived exosomal miRNA-26a-5p induces osteogenic differentiation of bone mesenchymal stem cells (133); Exosomal miR-486-5p secreted by M2 macrophage influences the differentiation potential of bone marrow mesenchymal stem cells and osteoporosis (134). These miRNAs enhance osteoblast differentiation and bone matrix deposition by inhibiting negative regulatory factors and activating key transcription factors, such as Runx2 and Osterix (135,136). Additionally, exosomal proteins such as TGF-β and BMPs support mineralization by regulating osteoblast proliferation and differentiation (137). TGF-β activates the Smad signaling pathway through receptor binding, enhancing osteoblast mineralization (138). BMPs, on the other hand, activate the Smad1/5/8 pathway, which promotes osteoblast differentiation and stimulates bone matrix synthesis and mineralization (139), as summarized in Table V (140-143).

Table V

Regulation of the proliferation, differentiation, and mineralization of osteoblasts.

Table V

Regulation of the proliferation, differentiation, and mineralization of osteoblasts.

Authors, yearActive moleculeMechanism of actionSignaling pathway/targetEffect(Refs.)
Méndez-Mancilla et al, 2024miR-21
miR-29
Promotes osteoblast proliferation and differentiation via pathway activationPI3K/Akt, Wnt/β-cateninEnhances osteoblast proliferation and differentiation(140)
Fu et al, 2018Runx2, OsterixmiRNA-mediated inhibition of negative regulators and activation of key transcriptionfactorsRunx2, OsterixFacilitates osteoblast differentiation and bone matrix deposition(141)
Luo et al, 2017TGF-βActivates Smad signaling through receptor bindingSmadImproves mineralization capacity of osteoblasts(142)
Zou et al, 2021BMPEnhances bone matrix synthesis and mineralization via Smad1/5/8 signalingSmad1/5/8Promotes osteoblast differentiation and bone matrix mineralization(143)

[i] miRNAs, microRNAs; Runx2, regulation of Runt-related transcription factor 2; wnt, wingless-type MMTV integration site family; Runx2, Runt-related transcription factor 2; TGF-β, transforming growth factor-beta; BMP, bone morphogenetic protein; Smad, small mothers against decapentaplegic; PI3K, phosphatidylinositol 3-kinase; AKT, protein kinase B.

Affecting the expression of osteoblast-related genes (such as Runx2 and Osterix)

The function of osteoblasts is further regulated by a series of transcription factors, with Runx2 and Osterix being two critical regulators of osteogenic differentiation (144). Macrophage-derived exosomes influence osteoblast function by modulating the expression of these transcription factors (145,146). Exosomes secreted by macrophages carry specific miRNAs, such as miR-124-3p and miR-146a, which regulate Runx2 and Osterix expression (147,148). For example, miR-224-5p targets the 3' UTR of Runx2, preventing its degradation and thereby promoting osteoblast differentiation (149); miR-6879-5p carried by M2 macrophage-derived exosomes increases Runx2 expression and promotes osteogenic differentiation and aerobic glycolysis in human periodontal ligament stem cells (hPDLSCs) via modulating TRIM26-mediated ubiquitination of pyruvate kinase M (PKM) (150); M2 macrophage exosomes carrying miRNA-26a-5p can induce osteogenic differentiation of bone marrow-derived stem cells to inhibit lipogenic differentiation by promoting the expression of RUNX-2 (151). Additionally, miR-664-3p promotes osteoblast differentiation and mineralization by regulating Osterix expression (152). As a key transcription factor downstream of Runx2, Osterix drives bone matrix formation and mineralization (147). As well as miRNAs, proteins such as TGF-β and BMP in macrophage-derived exosomes also promote osteoblast differentiation by modulating Runx2 and Osterix expression (153,154). TGF-β enhances Runx2 expression through the activation of the Smad2/3 signaling pathway, promoting osteoblast differentiation (136). BMPs stimulate Osterix expression and enhance bone matrix deposition and mineralization via the Smad1/5/8 signaling pathway (155).

The effect on osteoclast function
Regulating the formation and activation of osteoclasts

The formation and activation of osteoclasts, the primary cells responsible for bone resorption, are essential processes in bone metabolism (156). Macrophage-derived exosomes contribute to bone resorption and regulate bone metabolism by modulating osteoclast formation and activation (157). Osteoclast formation is governed by various factors, including Receptor Activator of Nuclear Factor κ-B Ligand (RANKL) and Macrophage Colony-Stimulating Factor (M-CSF) (158,159). Macrophage-derived exosomes promote osteoclast formation by carrying RANKL and M-CSF (160,161). Specifically, exosomal RANKL binds to the RANK receptor on osteoclasts, activating the NF-κB and MAPK signaling pathways to drive osteoclast formation and activation (162,163). By binding to its receptor c-Fms, M-CSF activates the PI3K/Akt signaling pathway, accelerating osteoclast differentiation (164). Additionally, macrophage-derived exosomes can further promote osteoclast formation by modulating other cytokines, such as IL-1 and TNF-α, which enhance RANKL expression through the activation of the NF-κB signaling pathway, thereby promoting osteoclast activation (57). Consequently, macrophage-derived exosomes facilitate bone resorption by carrying RANKL, M-CSF, and other factors that regulate osteoclast formation and activation.

Changing the molecular mechanism related to osteoclast bone resorption activity

Osteoclast bone resorption activity is critical for regulating bone metabolism (165). Macrophage-derived exosomes modulate bone metabolism by influencing osteoclast resorption activity and altering underlying molecular mechanisms (166). The bone-resorbing capacity of osteoclasts heavily depends on the activation of surface receptors, particularly the RANKL/RANK ligand-receptor interaction (167). Macrophage-derived exosomes carrying RANKL can activate RANK receptors on osteoclasts, enhancing bone resorption activity (168). Moreover, exosomal matrix metalloproteinases (MMPs) contribute to bone resorption by degrading the bone matrix (169). Therefore, macrophage-derived exosomes augment osteoclast activity and bone resorption not only through the RANKL-RANK axis but also via MMPs and other molecules. Exosomal miRNAs and protein factors further regulate the molecular mechanisms of bone resorption by modulating enzymatic activities within osteoclasts (170). For instance, miR-146a promotes osteoclast activity by targeting negative regulators of the NF-κB pathway. By contrast, TGF-β enhances bone resorption by activating the Smad signaling pathway in osteoclasts (171). In summary, macrophage-derived exosomes play a pivotal role in regulating bone metabolism by modulating the molecular pathways that control osteoclast-mediated bone resorption.

Function in pathological state

IGF-1 and FGF-2 play pivotal roles in both bone and muscle metabolism. Osteoporosis, often accompanied by SP, is a condition that involves diminished bone density and function, alongside muscle degradation. Macrophage-derived extracellular vesicles have been shown to improve both osteoporosis and SP by regulating IGF-1 and FGF-2 signaling.

Osteoporosis
Changes of macrophage-derived exosomes, IGF-1 and FGF-2 in the pathogenesis of osteoporosis

Osteoporosis is characterized by reduced bone density, deterioration of bone microarchitecture and increased fracture risk, primarily affecting older adults (172). Recent studies suggest that osteoporosis is not only associated with abnormal bone metabolism but also with impaired muscle function (Figs. 11 and 12), highlighting the significance of musculoskeletal crosstalk in the pathogenesis of osteoporosis (Figs. 11 and 12) (172,173). In osteoporosis, the functions of macrophage-derived exosomes, IGF-1, and FGF-2 are markedly altered. Macrophages, as key immune cells (174,175), contribute to bone metabolism through exosomes that transport a variety of signaling molecules, including cytokines, miRNAs, and proteins (176). Impaired macrophage function in patients with osteoporosis alters the signaling molecule profile within exosomes (177). These changes may accelerate bone loss by disrupting the balance between bone resorption and formation through various signaling pathways. IGF-1 plays a central role in bone metabolism by regulating bone formation, primarily through the PI3K/Akt and MAPK signaling pathways (178,179).

Regulatory mechanism of estrogen
deficiency on bone metabolism during osteoporosis. Estrogen
deficiency leads to reduced muscle synthesis and increased protein
breakdown, resulting in muscle atrophy. Specifically, it decreases
IGF-1 synthesis, inhibiting the PI3K/Akt/mTOR pathway and reducing
muscle fiber protein synthesis. Concurrently, elevated MuRF1/MAFbx
activity promotes muscle degradation. Impaired PGC-1α function
reduces mitochondrial biogenesis, electron transport chain activity
and ATP production. Increased PINK1/Parkin-mediated mitophagy
clears dysfunctional mitochondria. Enhanced M1 macrophage activity
contributes to muscle tissue inflammation. PI3K,
phosphatidylinositol-3-kinase; Akt, protein kinase B; MuRF1, muscle
RING finger-1; IGF-1, insulin-like growth factor 1; mTOR, mammalian
target of rapamycin; PGC-1α, Peroxisome proliferator-activated
receptor γ coactivator 1α; MAFbx, F-box only protein 32; PINK1,
PTEN-induced putative kinase 1; ATP, Adenosine triphosphate; Bax,
Bcl-2-associated X protein; BAK, BCL-2 antagonist killer 1.

Figure 11

Regulatory mechanism of estrogen deficiency on bone metabolism during osteoporosis. Estrogen deficiency leads to reduced muscle synthesis and increased protein breakdown, resulting in muscle atrophy. Specifically, it decreases IGF-1 synthesis, inhibiting the PI3K/Akt/mTOR pathway and reducing muscle fiber protein synthesis. Concurrently, elevated MuRF1/MAFbx activity promotes muscle degradation. Impaired PGC-1α function reduces mitochondrial biogenesis, electron transport chain activity and ATP production. Increased PINK1/Parkin-mediated mitophagy clears dysfunctional mitochondria. Enhanced M1 macrophage activity contributes to muscle tissue inflammation. PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; MuRF1, muscle RING finger-1; IGF-1, insulin-like growth factor 1; mTOR, mammalian target of rapamycin; PGC-1α, Peroxisome proliferator-activated receptor γ coactivator 1α; MAFbx, F-box only protein 32; PINK1, PTEN-induced putative kinase 1; ATP, Adenosine triphosphate; Bax, Bcl-2-associated X protein; BAK, BCL-2 antagonist killer 1.

Regulatory mechanism of estrogen
deficiency on muscle metabolism during osteoporosis. Estrogen
deficiency disrupts bone remodeling by decreasing PGC-1α and
elevating SOST and FSH, leading to increased inflammatory cytokines
(such as IL-1, IL-6, IL-17 and TNF-α) and oxidative stress. These
changes promote osteoclast differentiation and enhance bone
resorption. Simultaneously, MSCs shift differentiation potential
toward adipogenesis at the expense of osteogenesis. This imbalance
between increased bone resorption and decreased bone formation
results in impaired bone remodeling and net bone loss. PGC-1α,
Peroxisome proliferator-activated receptor γ coactivator 1α; SOST,
Sclerostin; FSH, Follicle-stimulating hormone; MSCs, Mesenchymal
stem cells.

Figure 12

Regulatory mechanism of estrogen deficiency on muscle metabolism during osteoporosis. Estrogen deficiency disrupts bone remodeling by decreasing PGC-1α and elevating SOST and FSH, leading to increased inflammatory cytokines (such as IL-1, IL-6, IL-17 and TNF-α) and oxidative stress. These changes promote osteoclast differentiation and enhance bone resorption. Simultaneously, MSCs shift differentiation potential toward adipogenesis at the expense of osteogenesis. This imbalance between increased bone resorption and decreased bone formation results in impaired bone remodeling and net bone loss. PGC-1α, Peroxisome proliferator-activated receptor γ coactivator 1α; SOST, Sclerostin; FSH, Follicle-stimulating hormone; MSCs, Mesenchymal stem cells.

Influence of the interaction between the three on bone loss

The primary manifestations of osteoporosis, bone loss and reduced bone strength, are influenced by the interaction of macrophage-derived exosomes, IGF-1 and FGF-2 (180). These components synergistically regulate bone formation and resorption, thereby affecting bone mass (181). Exosomal miRNAs, such as miR-21 and miR-146a, derived from macrophages, can promote osteoclast differentiation and activity by modulating the RANKL/RANK pathway, leading to excessive bone resorption (182). Additionally, exosomal protein factors such as TGF-β and BMP further inhibit bone formation by regulating osteoblast function (183). In osteoporosis, IGF-1, a key osteogenic factor, is often underexpressed, impairing osteogenesis and leading to reduced bone formation (184). Similarly, FGF-2 signaling is suppressed in osteoporosis, contributing to further bone density loss (185). This disruption in signaling between IGF-1 and FGF-2 exacerbates bone loss. Exosomes carrying RANKL and TGF-β promote osteoclastogenesis and bone resorption (186), while the deficiency of IGF-1 and FGF-2 signaling further impairs bone formation. Disruption of this delicate musculoskeletal crosstalk and the associated molecular pathways plays a pivotal role in the pathogenesis of osteoporosis (187).

Abnormal bone metabolism associated with sarcopenia
Association between muscle atrophy and changes in bone metabolism in sarcopenia

SP refers to the age-related progressive loss of skeletal muscle mass and function (188). It is not only a natural part of aging but is also closely linked to various diseases and pathological conditions. The relationship between SP and abnormal bone metabolism has gained considerable attention in recent years. Muscle atrophy and bone metabolism interact bidirectionally, forming a critical regulatory mechanism for maintaining bone health (189). As depicted in Fig. 13, the onset of SP is often accompanied by decreased bone density and altered bone metabolism. Muscle atrophy influences bone metabolism through multiple mechanisms (190). It reduces muscle strength and load-bearing capacity, thereby diminishing mechanical stress on bones. This reduction in mechanical stimulation leads to increased bone resorption and decreased bone formation (190). Additionally, SP affects bone metabolism through the secretion of inflammatory factors, such as IL-6 and TNF-α (191). These inflammatory mediators activate osteoclast signaling pathways, enhancing bone resorption and consequently reducing bone density (192). Moreover, endocrine dysfunction linked to SP plays a significant role in abnormal bone metabolism. Muscle atrophy leads to reduced secretion of myokines, such as IGF-1 and FGF-2, impairing bone formation.

Regulatory mechanism of sarcopenia on
bone metabolism during osteoporosis. Sarcopenia, characterized by
the loss of muscle mass and reduced mechanical loading, leads to
decreased osteogenic activity and bone formation. Impaired muscle
contraction reduces Wnt/β-catenin signaling, inhibiting osteoblast
differentiation. Declines in IGF-1 synthesis suppress PI3K/Akt/mTOR
activity, further impairing osteoblast differentiation. Increased
inflammatory cytokines elevate RANKL expression and suppress OPG,
enhancing osteoclast activity and bone resorption. Reduced vitamin
D intake decreases intestinal calcium absorption and bone
formation. Muscle atrophy triggers a stress response, increasing
norepinephrine production and sustained β2-AR activation, which
promotes adipogenic differentiation of bone marrow mesenchymal stem
cells. Muscle mitochondrial dysfunction reduces ATP synthesis,
inhibits SERCA activity, causes intracellular calcium overload, and
induces osteoblast apoptosis. PI3K, phosphatidylinositol-3-kinase;
Akt, protein kinase B; IGF-1, insulin-like growth factor 1; mTOR,
mammalian target of rapamycin; ATP, Adenosine triphosphate; RANKL,
receptor activator of nuclear factor-κB ligand; OPG,
osteoprotegerin.

Figure 13

Regulatory mechanism of sarcopenia on bone metabolism during osteoporosis. Sarcopenia, characterized by the loss of muscle mass and reduced mechanical loading, leads to decreased osteogenic activity and bone formation. Impaired muscle contraction reduces Wnt/β-catenin signaling, inhibiting osteoblast differentiation. Declines in IGF-1 synthesis suppress PI3K/Akt/mTOR activity, further impairing osteoblast differentiation. Increased inflammatory cytokines elevate RANKL expression and suppress OPG, enhancing osteoclast activity and bone resorption. Reduced vitamin D intake decreases intestinal calcium absorption and bone formation. Muscle atrophy triggers a stress response, increasing norepinephrine production and sustained β2-AR activation, which promotes adipogenic differentiation of bone marrow mesenchymal stem cells. Muscle mitochondrial dysfunction reduces ATP synthesis, inhibits SERCA activity, causes intracellular calcium overload, and induces osteoblast apoptosis. PI3K, phosphatidylinositol-3-kinase; Akt, protein kinase B; IGF-1, insulin-like growth factor 1; mTOR, mammalian target of rapamycin; ATP, Adenosine triphosphate; RANKL, receptor activator of nuclear factor-κB ligand; OPG, osteoprotegerin.

The role of macrophage-derived exosomes, IGF-1 and FGF-2 in this association

Macrophage-derived exosomes, along with IGF-1 and FGF-2, play pivotal roles in the bone metabolism abnormalities associated with SP (193,194). Macrophages, as key immune cells, secrete exosomes carrying a range of cytokines and miRNAs that are essential for musculoskeletal crosstalk (195). The miRNAs, cytokines and growth factors transported by macrophage exosomes affect all aspects of bone metabolism (196). For example, macrophage-derived exosomes promote osteoclast formation and activity by delivering miRNAs (such as miR-146a and miR-21), thus enhancing bone resorption (197). These exosomal miRNAs can activate specific signaling pathways by binding to receptors on bone cells, influencing the pathogenesis of conditions such as osteoporosis and bone loss (197). Moreover, the roles of IGF-1 and FGF-2 in SP are particularly important (198). IGF-1 is well-established as essential for muscle tissue, promoting not only muscle growth and repair but also bone matrix formation by enhancing osteoblast function (199). However, circulating IGF-1 levels are often reduced in patients with SP, contributing to bone metabolism disorders. Similarly, FGF-2 is another key osteogenic factor involved in SP.

Research methods and technical means

Exosome isolation from macrophages typically involves techniques such as ultracentrifugation, size exclusion chromatography, immunoaffinity capture, and kit-based methods. Identification techniques include transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), western blotting (WB) and nanoflow cytometry. Each method offers distinct advantages and limitations. In muscle and bone metabolism experiments, in vitro methods include osteoblast differentiation assays, osteoclast inhibition tests, muscle cell differentiation experiments, and muscle metabolism studies. In vivo approaches include osteoporosis models, fracture healing models, and SP models.

Separation and identification of exosomes
Comparison and optimization of exosome separation techniques such as the ultrafast centrifugal method and the kit method

Exosomes are nanoscale extracellular vesicles (30-150 nm in diameter) secreted by cells and are widely distributed in various body fluids (200). The isolation and purification of exosomes from macrophages is a critical step in studying musculoskeletal crosstalk and its impact on bone metabolism (201). Conventional methods for exosome isolation primarily include ultracentrifugation and commercial kit-based approaches (201). Ultracentrifugation is regarded as the gold-standard technique for exosome separation, relying on density-based differentiation to isolate exosomes from other cellular components (202). This process involves removing cellular debris through low-speed centrifugation, followed by high-speed ultracentrifugation to pellet exosomes (202). Although this method is widely adopted, straightforward and cost-effective, it is time-consuming and requires significant technical expertise (202). Despite these limitations, ultracentrifugation remains the most commonly used method due to its high yield and efficiency in exosome collection (203). By contrast, commercial kit-based methods use reagents for exosome separation, often employing immunomagnetic beads or affinity capture technologies (204). These kits are convenient, easy to use, and improve exosome purity markedly (204). However, they tend to be expensive and may exhibit lower extraction efficiency, especially in samples with high levels of contaminating cellular material (204). A comparative summary of these techniques is provided in Tables VI and VII (205-216).

Table VI

Comprehensive overview of exosome isolation techniques.

Table VI

Comprehensive overview of exosome isolation techniques.

Authors, yearMethodPrincipleApplication scenariosAdvantagesLimitations(Refs.)
Coughlan et al, 2020 UltracentrifugationSeparates exosomes based on density and size using high-speed centrifugation (100,000-200,000 × g)Bulk exosome preparation; proteomic/lipidomic analysisHigh yield; no chemical additives requiredTime-consuming (4-6 h); potential vesicle damage(205)
Sidhom et al, 2020Size-Exclusion ChromatographyPorous beads retain larger particles; exosomes elute firstClinical studies; therapeutic developmentHigh purity; maintains vesicle integrityLow throughput; requires sample pre-filtration(206)
Gao et al, 2022Polymer precipitation (such as PEG)Polymers reduce exosome solubility, inducing precipitationLarge-volume processing; initial screeningSimple protocol; cost-effectiveLow purity (co-precipitates contaminants); polymer carryover(207)
Greening et al, 2015Immunoaffinity captureUses antibodies against surface markers (such as CD9, CD63, CD81)Biomarker discovery; targeted isolationHigh specificity; suitable for rare exosomesExpensive (>$500/sample); antibody-dependent bias(208)
Contreras-Naranjo et al, 2017MicrofluidicsSize- or charge-based sorting through nanochannelsPoint-of-care diagnostics; small sample volumesRapid (30-60 min); portable systems availableLow yield (<50%); requires specialized equipment(209)
Tang et al, 2017Kit methodUtilizes immunomagnetic beads or affinity capture for isolationHigh-purity exosome extraction from complex samplesImproved efficiency and purityHigher cost; more complex operation(210)
Huang et al, 2022UC + kit methodCombines ultracentrifugation and kit-based strategies to mitigate limitations of eachApplications requiring balanced extraction efficiency and purityEnhanced separation efficiency and purityIncreased cost and operational complexity(211)

Table VII

Top 5 exosome isolation kits.

Table VII

Top 5 exosome isolation kits.

Authors, yearNameMethodSample CompatibilityTimeYieldPurityPrice (US$)(Refs.)
Huang et al, 2021ExoQuick-TC™ (SBI)Polymer precipitationSerum, plasma, urine30 minHighModerate200-400(212)
Lai et al, 2022qEV2® (Izon Science)Size-exclusion chromatographyCell culture, CSF1 hMediumHigh300-600(213)
D'Acunzo et al, 2022Total Exosome Isolation™ (Thermo Fisher Scientific, Inc.)Polymer-based precipitationBiofluids, cell media30 minHighLow250-450(214)
Doyle et al, 2019Exo-FETCH™ (Bio-Techne)Anti-CD63/CD81 immunoaffinitySmall volumes (50-200 μl)2 hLowVery high500-800(215)
Veerman et al, 2021ExoEasy Maxi™ (Qiagen GmbH)Membrane affinityLarge volumes (up to 4 ml)1.5 hHighMedium400-700(216)

[i] CSF, cerebrospinal fluid.

Identification methods of exosomes (such as electron microscopy, WB detection of marker proteins)

Exosome identification is a key aspect of exosome research (217). Commonly used characterization methods include electron microscopy, WB, and NTA (217). Electron microscopy is the most traditional approach for morphological characterization, allowing direct visualization of exosomes as spherical vesicles (212). Scanning electron microscopy (SEM) and TEM enable precise determination of exosome size, shape, and membrane structure (218). WB is routinely employed to identify exosomes by detecting specific marker proteins, including CD9, CD63, TSG101, and Alix (219). These proteins are highly enriched on the exosomal membrane and serve as canonical markers for exosome identification (219). Additionally, WB can analyze various exosomal cargoes (such as proteins, receptors and enzymes), offering insights into their biological functions in musculoskeletal crosstalk (220). NTA measures the size distribution of exosomes based on their Brownian motion, with most exosomes ranging from 30 to 150 nm in diameter (221).

Experimental cell model
Establishment of a co-culture system of muscle cells and bone cells

Investigating the impact of macrophage-derived exosomes, IGF-1, and FGF-2 on musculoskeletal crosstalk requires the establishment of a co-culture system that incorporates both muscle cells and osteocytes. Commonly used bone cell models include the MC3T3-E1 murine pre-osteoblastic cell line and the RAW264.7 murine monocytic cell line (222). These cell lines exhibit high proliferative and differentiation capacities, making them ideal for in vitro investigations (222). For muscle cell models, the C2C12 mouse myoblast cell line and the L6 rat skeletal muscle cell line are frequently used (223). The C2C12 cell line, in particular, differentiates into myotubes under appropriate conditions and is widely employed in muscle biology research (224). This co-culture model simulates the in vivo physiological environment and serves as a robust platform for studying muscle-bone interactions. Co-culture systems are typically established using either Transwell inserts or direct contact methods. In the Transwell system, a porous membrane (typically 0.4 μm) separates the cell types, enabling paracrine signaling via soluble factors while preventing direct cell-cell contact (225). This setup allows for precise control over the transfer of soluble factors between cell compartments (225). However, direct contact co-culture more accurately replicates the physical interactions and signaling processes, including mechanical stimulation, between muscle and bone cells (226).

Experimental designs to study the interaction of macrophage-derived exosomes, IGF-1 and FGF-2 in a cell model

Experimental designs investigating the interactions between macrophage-derived exosomes, IGF-1, and FGF-2 typically involve four key components: Exosome isolation and processing, gene interference, protein expression analysis and functional assays. A critical step in this experimental design is the processing of exosomes, typically secreted by macrophage cell lines such as RAW264.7. Exosomes from M1 macrophages are enriched with pro-inflammatory factors, while exosomes derived from M2 macrophages carry anti-inflammatory and tissue-repair factors (227). In experiments, exosomes isolated from macrophage cell lines (such as RAW264.7) are introduced into muscle and bone cell co-culture systems to assess their effects on cellular proliferation, differentiation, and mineralization (228). To determine the specific influence of IGF-1 and FGF-2, their functions can be inhibited using specific antagonists or gene interference techniques, such as siRNA, to confirm their roles in mediating musculoskeletal crosstalk. Additionally, techniques such as WB and qPCR can be employed to analyze how M1 and M2 exosomes regulate IGF-1 and FGF-2 expression, as well as the activity of their downstream signaling pathways, including PI3K/Akt and MAPK (229). Functional assays, such as osteogenic evaluation via Alizarin Red staining and osteoclastic activity assessment using TRAP staining, can be used to verify the effects of exosomes on bone cell function (230). Collectively, this experimental approach provides a framework for elucidating the complex interactions and regulatory mechanisms involving M1- and M2-macrophage-derived exosomes, IGF-1, and FGF-2 in bone metabolism.

Animal experimental model
Selection of suitable animal models (such as mice and rats) to simulate bone metabolism-related diseases

As summarized in Table VIII (231-239), animal models form the foundation for studying bone metabolism and related diseases. Commonly used mouse and rat models effectively simulate bone metabolic disorders and offer insights into the molecular mechanisms underlying bone metabolism; however, each model has distinct advantages and limitations. In osteoporosis research, ovariectomized mouse or rat models are frequently used to mimic the disruption of bone metabolism caused by estrogen deficiency (240). Additionally, drug interventions or genetic knockout techniques can establish disease models related to bone metabolism, allowing the investigation of the effects of exosomes, IGF-1, and FGF-2 (241,242). Bilateral ovariectomy is widely used to model postmenopausal osteoporosis (243), recapitulating key osteoporosis features, such as reduced bone mass and density (244). In rat models, glucocorticoid administration (such as dexamethasone) is commonly employed to induce abnormal bone metabolism, simulating drug-induced osteoporosis (245). These animal models provide valuable platforms for investigating the role of exosomes in bone metabolism.

Table VIII

Animal models for bone metabolism research.

Table VIII

Animal models for bone metabolism research.

Authors, yearDiseaseModelInduction methodAdvantagesLimitationsBest applications(Refs.)
Tenkumo et al, 2020OsteoporosisOVX mice/ratsOvariectomy (estrogen loss)Recapitulates human pathophysiologyLong modeling time (3-6 months)Evaluating anti-resorptive therapies(231)
Halloran et al, 2024Aged C57BL/6 miceNatural aging (≥18 months)Spontaneous developmentHigh individual variabilityStudying age-related bone loss(232)
Yashima et al, 2025Fracture HealingSD rat drill-holeSurgical defect (1-2 mm)Quantifiable healing processRequires micro-CT monitoringBiomaterial and scaffold evaluation(233)
Muwanga et al, 2022Mouse tibial fractureOpen fracture with fixationClinically relevant modelTechnically challengingMechanical loading studies(234)
Wang et al, 2019OsteoarthritisDMM mouse/ratMedial meniscus destabilizationProgressive cartilage degenerationSlow progression (8-12 weeks)Disease-modifying drug testing(235)
Suh et al, 2022MIA ratSodium iodoacetate injectionRapid induction (1-2 weeks)Prominent acute inflammationAnti-inflammatory agent screening(236)
Li et al, 2024Bone TumorsNude mouse OS modelIntratibial MG63 cell injectionHuman-like tumor microenvironmentLacks immune componentMetastasis/therapy studies(237)
Ferrena et al, 2024p53-KO transgenic mouseSpontaneous osteosarcomaGenetically relevantUnpredictable tumor onsetTumorigenesis mechanisms(238)
Wang et al, 2024Metabolic Bone DiseaseHFD+STZ diabetic ratHigh-fat diet + streptozotocinModels diabetic osteopeniaVariable glycemic controlGlucose-bone interaction studies(239)

[i] OVX, ovariectomy; micro-CT, micro-computed tomography; SD, Sprague Dawley; p53 KO, p53 knock-out; OS, open source; DMM, destabilized medial meniscus; MIA, monosodium iodoacetate; MG63 cells, osteoblast-like osteosarcoma cells; HFD, high-fat diet; STZ, streptozotocin.

Intervention methods and detection indicators of macrophage-derived exosomes, IGF-1, and FGF-2 in animal experiments

In animal studies, various intervention strategies are employed to investigate the effects of macrophage-derived exosomes (from M1 or M2 phenotypes), IGF-1, FGF-2 and related factors on bone metabolism. These agents, such as M1- or M2-derived exosomes, IGF-1 and FGF-2, are typically administered through local injection or systemic intravenous delivery. Researchers establish multiple experimental groups, including those treated with M1-derived exosomes, M2-derived exosomes, IGF-1, FGF-2, and vehicle controls, to enable direct comparison of the specific effects of each intervention on bone metabolism. Standard detection endpoints include bone densitometry such as dual-energy X-ray absorptiometry, histological analysis of bone tissue (such as hematoxylin and eosin [H&E] staining, Alizarin Red staining) and serum biomarkers of bone metabolism (such as osteocalcin, C-terminal telopeptide of type I collagen, bone-specific alkaline phosphatase) (246). These parameters offer a comprehensive evaluation of bone metabolism, allowing for an in-depth assessment of the effects mediated by M1 and M2 macrophage-derived exosomes. Collectively, these animal experiments shed light on how M1 and M2 macrophage-derived exosomes modulate IGF-1 and FGF-2 signaling within the musculoskeletal crosstalk network, influencing osteocyte proliferation, differentiation and mineralization. These findings provide a foundation for developing novel therapeutic strategies.

Effects of external factors on the osteogenic function of macrophage-derived exosomes

Diabetes increases the risk of delayed fracture healing and nonunion (247). Chen et al (242) found that alterations in the diabetic microenvironment induce functional changes in macrophages, leading to exosomes with markedly impaired osteogenic capacity when interacting with BMMSCs, compared with those from normal macrophages. In vitro assays demonstrate reduced mineralization and significant downregulation of osteogenic genes, consistent with in vivo findings (248). High blood sugar, insulin resistance, and inflammatory factors (such as TNF-α and IL-6) promote an increase in pro-inflammatory M1 macrophages (249). These macrophages secrete exosomes rich in pro-inflammatory miRNAs (such as miR-155, miR-214 and miR-146) and inflammatory factors (250). For instance, miR-155-containing exosomes inhibit the Wnt/β-catenin signaling pathway, reducing osteoblast differentiation (251). M1 macrophage-derived exosomes, rich in pro-inflammatory factors like TNF-α and IL-6, activate the NF-κB pathway, inhibit Runx2 expression and suppress osteoblast differentiation (252). Moreover, M1 macrophage-driven exosomal miR-214 inhibits OPG, leading to an imbalance in the RANKL/OPG ratio and accelerating bone loss (253). Additionally, exosomal miR-146 inhibits IRS-1/Akt signaling, exacerbating insulin-resistant muscle atrophy (122). High blood sugar induces reactive oxygen species (ROS) accumulation, altering the protein and nucleic acid composition of macrophage-derived exosomes, including the presence of oxidative damage markers such as 8-OHdG (254). M1 exosomes containing ROS/glycation end-products (AGEs) can impair mitochondrial function, reduce ATP production and contribute to muscle weakness (255). Similarly, Song and Chung (256) demonstrated that exosomes from Porphyromonas gingivalis-infected macrophages inhibit MC3T3-E1 osteoblast proliferation, promote apoptosis, downregulate osteogenic gene expression and suppress osteogenic differentiation. Liu et al (257) observed that zinc ion concentration differentially modulates the osteogenic function of macrophage-derived exosomes. At 4 μmol/l ZnCl2, macrophage-derived exosomes exhibited the strongest pro-osteogenic effects on osteoblasts, while at 20 μmol/l, they enhanced endothelial cell migration. Zhu et al (258) reported that magnesium ions (Mg2+) promote macrophage autophagy and polarization, enhancing the pro-osteogenic effects of their exosomes on BMMSCs. Wei et al (259) showed that exosomes from BMP-2-stimulated macrophages markedly enhance osteogenesis. The osteogenic capacity of macrophage-derived exosomes is clearly influenced by external factors (260). Further research is needed to identify additional factors that modulate the osteogenic capacity of macrophage-derived exosomes, which holds important clinical implications.

The effect of sex differences on the role of estrogen in bone metabolism and muscle metabolism

Estrogen influences bone and muscle metabolism differently in males and females (Table IX) (261-271). The decline in estrogen levels is the primary cause of postmenopausal osteoporosis in women (240), leading to reduced bone mass, muscle atrophy and an increased risk of falls and fractures (240). SP often coexists with osteoporosis, resulting in osteosarcopenia (272). While male androgens predominantly regulate muscle and bone metabolism, estrogen remains crucial for maintaining bone strength and muscle metabolic adaptability (267). Further studies indicate that in obesity or infection models, male macrophages tend to be M1 polarized, with their exosomes rich in pro-inflammatory miRNAs (such as miR-155) that inhibit the insulin signaling pathway (273). By contrast, macrophage extracellular vesicles in female models carry higher levels of anti-inflammatory factors (such as miR-125a-5p), enhancing tissue repair (274). The decrease in estrogen levels in postmenopausal women leads to an M1/M2 imbalance, accelerating bone loss (with osteoporosis rates twice as high as in men) (274). In males, high testosterone levels are positively associated with muscle mass (275). However, obese males exhibit higher expression of miR-155 in macrophage exosomes from adipose tissue, which increases the risk of insulin resistance (276). M2 macrophage exosomes mediate the polarization of macrophages into anti-inflammatory phenotypes in female models, accelerating muscle regeneration (277). These findings suggest that estrogen exerts distinct effects on bone and muscle metabolism across sexes, leading to different regulatory mechanisms of macrophage exosomes in estrogen-induced muscle-bone metabolism abnormalities, influenced by sex-specific factors.

Table IX

Estrogen's key roles in bone density and muscle metabolism, sex-specific summary.

Table IX

Estrogen's key roles in bone density and muscle metabolism, sex-specific summary.

Authors, yearFunctionEffects in FemalesEffects in malePrimary mechanismClinical relevance(Refs.)
Moura et al, 2018; Vilaca et al, 2022; Cauley 2015Bone Density Regulation• Maintains bone mass by inhibiting osteoclast activity (reducing bone resorption).
• Promotes osteoblast proliferation and bone formation.
• Rapid bone loss post-menopause due to estrogen decline (↑ fracture risk).
• Essential for maintaining bone mineralization via ERα/ERβ receptors.
• Deficiency disrupts bone metabolism, leading to osteoporosis.
• Aromatase converts testosterone to estrogen, indirectly protecting bones.
• Binds to estrogen receptors (ERα/ERβ) on bone cells.
• Suppresses RANKL pathway, ↑ osteoprotegerin (OPG) to inhibit osteoclasts.
• Enhances calcium absorption via vitamin D activation.
• HRT reduces postmenopausal osteoporosis risk.
• SERMs (such as Raloxifene) mimic estrogen's bone-protective effects.
(261-263)
Pellegrino et al, 2022; Alexander et al, 2022; Jardí et al, 2018Muscle metabolism• Preserves muscle mass by reducing protein degradation.
• Declines post-menopause contribute to sarcopenia (muscle loss) and ↓ strength.
• Indirectly supports muscle function via mitochondrial stability.
• ESR1/ERα remodels mitochondrial cristae-nucleoid structure, ↑ metabolic resilience.
• Enhances exercise adaptation and insulin sensitivity in skeletal muscle.
• Low estrogen ↑ fat accumulation, ↓ muscle quality.
• Modulates mitochondrial biogenesis and oxidative metabolism.
• Regulates insulin/IGF-1 signaling pathways.
• Anti-inflammatory effects protect against muscle atrophy.
• Estrogen-based therapies may prevent sarcopenia in aging.
• ERα activation counters metabolic diseases in males.
(264-266)
Unger et al, 2023; Collins et al, 2018; Tramunt et al, 2020; O'Reilly et al, 2021; Farhat et al, 2017Key differencesEstrogen deficiency directly drives accelerated bone/muscle loss post-menopause.Effects are subtler but critical for metabolic flexibility and musculoskeletal health.Females: Dominant via direct receptor binding. Males: Relies on local aromatization of testosterone.Screening for estrogen deficiency is vital in male osteoporosis.(267-271)

[i] IGF-1, insulin-like growth factor 1; ERα, estrogen receptor alpha; ERβ, estrogen receptor beta; SERMs; Selective estrogen receptor modulators; ESR1, estrogen receptor 1; OPG, osteoprotegerin; RANKL, receptor activator of NF-kappa B ligand; HRT, hormone replacement therapy; ERβ, estrogen receptor beta; SERMs, selective estrogen receptor modulators.

Challenge on macrophage exosomes in regulating musculoskeletal metabolism

Although RAB-GTPase-modified exosomes (RAB-EXOs), engineered via click chemistry, can target bone tissue in complex in vivo environments, their targeting and enrichment efficiencies remain suboptimal (278). Drug concentrations at sites of deep-seated bone infections, such as osteomyelitis, are often insufficient and systemic administration can result in off-target effects and potential adverse reactions. Studies show that intravenously injected exosomes are primarily taken up by the liver, skeletal muscle and adipose tissue (279-281). Enhancing exosome enrichment at specific bone lesion sites remains a critical challenge that needs urgent resolution (282). Exosomes derived from macrophages of different sources and polarization states exhibit significant differences in composition and biological function. Exosomes from M1 and M2 macrophages can exert opposing biological effects, and this heterogeneity presents a considerable challenge for developing standardized therapies. One study demonstrated that exosomes secreted by adipose tissue macrophages in obese mice promote insulin resistance (283), whereas those from M2 macrophages improve insulin sensitivity (284). Ensuring batch-to-batch consistency and functional stability of therapeutic exosomes is a major challenge, as is overcoming technical bottlenecks in their large-scale production. Exosomes isolated from 20-25 lean mice are needed to treat one obese mouse, yielding too little for clinical translation. Although in vitro induction of M2 macrophages can partially address source limitations, optimization of culture conditions, purification protocols and storage stability remains necessary. The use of composite technologies with carrier materials (such as hydrogels) also faces challenges in large-scale production. While specific miRNAs, such as miR-690, mediate the insulin-sensitizing effects of M2 macrophage-derived exosomes (284) and the PI3K/AKT pathway regulates macrophage polarization, significant knowledge gaps remain regarding their application in treating musculoskeletal tissues. The full molecular network underlying bone metabolic diseases is not yet fully understood (285). Key questions concerning exosomal release kinetics, interactions with host cells, and long-term effects of exosomal cargo require further in-depth investigation. This lack of knowledge hinders the precise design and optimization of therapeutic regimens.

Exosomes are nanoscale extracellular vesicles with diverse bioactivities; however, their long-term safety profile remains incompletely characterized. Although animal studies have shown that RAB-EXOs induce no adverse reactions within a 28-day period in osteomyelitis treatment (286), their immunogenicity, potential toxicity and the risk of off-target effects in human applications require systematic evaluation. This is especially important for patients with metabolic diseases, as exosome-based therapies may have complex and unpredictable effects on systemic metabolic networks. Consequently, a comprehensive safety evaluation framework and robust risk mitigation strategies must be established.

Conclusion

In conclusion, exosomes derived from M1 and M2 macrophages play pivotal roles in muscle-bone crosstalk, particularly within molecular signaling pathways mediated by myokines such as IGF-1 and FGF-2. These factors are essential regulators of muscle growth and bone metabolism, promoting muscle cell proliferation and differentiation while influencing bone cell function through various signaling pathways. Their roles extend beyond direct regulation of local cellular behavior, modulating bone metabolic homeostasis via macrophage-derived exosomes. Ongoing research continues to uncover the complex mechanisms through which M1 and M2 macrophage-derived exosomes affect bone metabolism. Exosomes facilitate intercellular communication by transporting bioactive molecules (such as proteins, RNAs and lipids), regulating bone tissue formation and remodeling. Specifically, exosomes secreted by M1 and M2 macrophages can modulate bone density and strength by influencing osteoblast and osteoclast activity, promoting bone health and aiding in repair. These findings provide valuable insights into the intricate mechanisms governing bone metabolism and lay the groundwork for developing new therapeutic strategies.

Future development

To address current challenges, researchers are exploring several strategies, including engineering targeted modification technologies to enhance exosomal tissue specificity, establishing standardized production and quality control protocols, applying multi-omics approaches to clarify mechanisms of action and developing smart, responsive carrier systems to improve delivery efficiency. Innovative solutions, such as M2 macrophage exosome-hydrogel composites, show promise for bone regeneration therapy, but further preclinical and clinical studies are essential to evaluate their safety and efficacy. Interdisciplinary collaboration will be vital in advancing this field. Further investigation into the mechanisms of M1 and M2 macrophage-derived exosomes is expected to lead to more effective interventions for bone-related diseases, including osteoporosis and fracture repair.

Availability of data and materials

Not applicable.

Authors' contributions

Conceptualization was by RMC, MZ, and JBH. Data curation was by ZXW and YFC. Funding acquisition was secured by MWL. Investigation was by SJG and YLZ. Project administration was performed by YLZ. Software management was overseen by ZBY. Figures 1-13 were prepared by MWL and RMC. Supervision was led by MWL. Validation was performed by RMC and visualization was by MZ. The original draft of the manuscript was written by MWL, who contributed to the review and editing. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Union Foundation of Yunnan Provincial Science and Technology Department and Kunming Medical University (grant no. 202201AY070001-091), Yunnan Province Clinical Center for Skin Immune Diseases (grant no. YWLCYXZX2023300076) and the Natural Science Foundation of China (grant no. 81960350).

References

1 

Kirk B, Lombardi G and Duque G: Bone and muscle crosstalk in ageing and disease. Nat Rev Endocrinol. 21:375–390. 2025. View Article : Google Scholar : PubMed/NCBI

2 

Shirvani H, Shamsoddini A, Bazgir B, McAinch AJ, Najjari A and Arabzadeh E: Metabolic crosstalk between skeletal muscle and cartilage tissue: Insights into myokines in osteoarthritis. Mol Biol Rep. 52:9572025. View Article : Google Scholar : PubMed/NCBI

3 

Chuang YH, Chuang WL, Huang SP and Huang CH: Expression of epidermal growth factor, basic fibroblast growth factor and insulin growth factor-1 and relation to myocyte regeneration of obstructed ureters in rats. Scand J Urol Nephrol. 39:7–14. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Cecerska-Heryć E, Goszka M, Serwin N, Roszak M, Grygorcewicz B, Heryć R and Dołęgowska B: Applications of the regenerative capacity of platelets in modern medicine. Cytokine Growth Factor Rev. 64:84–94. 2022. View Article : Google Scholar

5 

Gries KJ, Zysik VS, Jobe TK, Griffin N, Leeds BP and Lowery JW: Muscle-derived factors influencing bone metabolism. Semin Cell Dev Biol. 123:57–63. 2022. View Article : Google Scholar

6 

Pieters BCH, Cappariello A, van den Bosch MHJ, van Lent PLEM, Teti A and van de Loo FAJ: Macrophage-derived extracellular vesicles as carriers of alarmins and their potential involvement in bone homeostasis. Front Immunol. 10:19012019. View Article : Google Scholar : PubMed/NCBI

7 

Chen Y, Liu H, He Y, Yang B, Lu W and Dai Z: Roles for exosomes in the pathogenesis, drug delivery and therapy of psoriasis. Pharmaceutics. 17:512025. View Article : Google Scholar : PubMed/NCBI

8 

Yue Y, Cao S, Cao F, Wei Y, Li A, Wang D, Liu P, Zeng H and Lin J: Unveiling research hotspots: A bibliometric study on macrophages in musculoskeletal diseases. Front Immunol. 16:15193212025. View Article : Google Scholar : PubMed/NCBI

9 

Zheng A, Liu H, Yin G and Xie Q: Macrophage-derived exosomes in autoimmune diseases: Mechanistic insights and therapeutic implications. Immunol Res. 73:1712025. View Article : Google Scholar : PubMed/NCBI

10 

Fan C, Wang W, Yu Z, Wang J, Xu W, Ji Z, He W, Hua D, Wang W, Yao L, et al: M1 macrophage-derived exosomes promote intervertebral disc degeneration by enhancing nucleus pulposus cell senescence through LCN2/NF-κB signaling axis. J Nanobiotechnology. 22:3012024. View Article : Google Scholar

11 

Wen Z, Li S, Liu Y, Liu X, Qiu H, Che Y, Bian L and Zhou M: An engineered M2 macrophage-derived exosomes-loaded electrospun biomimetic periosteum promotes cell recruitment, immunoregulation, and angiogenesis in bone regeneration. Bioact Mater. 50:95–115. 2025.PubMed/NCBI

12 

Liu M, Ng M, Phu T, Bouchareychas L, Feeley BT, Kim HT, Raffai RL and Liu X: Polarized macrophages regulate fibro/adipogenic progenitor (FAP) adipogenesis through exosomes. Stem Cell Res Ther. 14:3212023. View Article : Google Scholar : PubMed/NCBI

13 

Zhou M, Li B, Liu C, Hu M, Tang J, Min J, Cheng J and Hong L: M2 Macrophage-derived exosomal miR-501 contributes to pubococcygeal muscle regeneration. Int Immunopharmacol. 101(Pt B): 1082232021. View Article : Google Scholar : PubMed/NCBI

14 

Yuan Z, Jiang D, Yang M, Tao J, Hu X, Yang X and Zeng Y: Emerging roles of macrophage polarization in osteoarthritis: Mechanisms and therapeutic strategies. Orthop Surg. 16:532–550. 2024. View Article : Google Scholar : PubMed/NCBI

15 

Ascenzi F, Barberi L, Dobrowolny G, Villa Nova Bacurau A, Nicoletti C, Rizzuto E, Rosenthal N, Scicchitano BM and Musarò A: Effects of IGF-1 isoforms on muscle growth and sarcopenia. Aging Cell. 18:e129542019. View Article : Google Scholar : PubMed/NCBI

16 

Park SH, Park J, Yoo JY, Kim HS, Lee M and Kim OK: Humulus japonicus enhances bone growth and microarchitecture in rats: Potential Involvement of IGF-1 signaling. J Med Food. 28:542–552. 2025.PubMed/NCBI

17 

Qiu Y, Yu B, Jiang C, Yin H, Meng J, Wang H, Chen L, Cai Y, Ren T, Qin Q, et al: Bone marrow mesenchymal stem cells overexpressing FGF-2 loaded onto a decellularized extracellular matrix hydrogel for the treatment of osteoarthritis. Biomater Sci. 14:9–30. 2026. View Article : Google Scholar

18 

Jaśkiewicz Ł, Romaszko-Wojtowicz A, Chmielewski G, Kuna J and Krajewska-Włodarczyk M: Effect of myokines on bone tissue metabolism: a systematic review. Bone. 201:1176542025. View Article : Google Scholar

19 

Zhao Z, Yan K, Guan Q, Guo Q and Zhao C: Mechanism and physical activities in bone-skeletal muscle crosstalk. Front Endocrinol (Lausanne). 14:12879722024. View Article : Google Scholar : PubMed/NCBI

20 

Yin L, Lu L, Lin X and Wang X: Crucial role of androgen receptor in resistance and endurance trainings-induced muscle hypertrophy through IGF-1/IGF-1R-PI3K/Akt-mTOR pathway. NutrMetab (Lond). 17:262020. View Article : Google Scholar

21 

Fu S, Yin L, Lin X, Lu J and Wang X: Effects of cyclic mechanical stretch on the proliferation of L6 myoblasts and its mechanisms: PI3K/Akt and MAPK signal pathways regulated by IGF-1 receptor. Int J Mol Sci. 19:16492018. View Article : Google Scholar : PubMed/NCBI

22 

Shen L, Li Y and Zhao H: Fibroblast growth factor signaling in macrophage polarization: Impact on health and diseases. Front Immunol. 15:13904532024. View Article : Google Scholar : PubMed/NCBI

23 

Zhang Z, Cao F, Liang D, Pan M, Lu WW, Lyu H, Xie Y, Zhang L and Tang P: Mechanical effects in aging of the musculoskeletal system: Molecular signaling and spatial scale alterations. J Orthop Translat. 52:464–477. 2025. View Article : Google Scholar : PubMed/NCBI

24 

Mao Y, Jin Z, Yang J, Xu D, Zhao L, Kiram A, Yin Y, Zhou D, Sun Z, Xiao L, et al: Muscle-bone cross-talk through the FNIP1-TFEB-IGF2 axis is associated with bone metabolism in human and mouse. Sci Transl Med. 16:eadk98112024. View Article : Google Scholar : PubMed/NCBI

25 

Yin P, Chen M, Rao M, Lin Y, Zhang M, Xu R, Hu X, Chen R, Chai W, Huang X, et al: Deciphering immune landscape remodeling unravels the underlying mechanism for synchronized muscle and bone aging. Adv Sci (Weinh). 11:e23040842024. View Article : Google Scholar

26 

Gómez-Bruton A, Matute-Llorente Á, González-Agüero A, Casajús JA and Vicente-Rodríguez G: Plyometric exercise and bone health in children and adolescents: A systematic review. World J Pediatr. 13:112–121. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Han J, Zhang J, Zhang X, Luo W, Liu L, Zhu Y, Liu Q and Zhang XA: Emerging role and function of Hippo-YAP/TAZ signaling pathway in musculoskeletal disorders. Stem Cell Res Ther. 15:3862024. View Article : Google Scholar : PubMed/NCBI

28 

Schiaffino S, Reggiani C, Akimoto T and Blaauw B: molecular mechanisms of skeletal muscle hypertrophy. J Neuromuscul Dis. 8:169–183. 2021. View Article : Google Scholar :

29 

Von den Hoff JW, Carvajal Monroy PL, Ongkosuwito EM, van Kuppevelt TH and Daamen WF: Muscle fibrosis in the soft palate: Delivery of cells, growth factors and anti-fibrotics. Adv Drug Deliv Rev. 146:60–76. 2019. View Article : Google Scholar

30 

Savadipour A, Palmer D, Ely EV, Collins KH, Garcia-Castorena JM, Harissa Z, Kim YS, Oestrich A, Qu F, Rashidi N and Guilak F: The role of PIEZO ion channels in the musculoskeletal system. Am J Physiol Cell Physiol. 324:C728–C740. 2023. View Article : Google Scholar : PubMed/NCBI

31 

Kirk B, Feehan J, Lombardi G and Duque G: Muscle, bone, and fat crosstalk: The biological role of myokines, osteokines, and adipokines. Curr Osteoporos Rep. 18:388–400. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Guo L, Quan M, Pang W, Yin Y and Li F: Cytokines and exosomal miRNAs in skeletal muscle-adipose crosstalk. Trends Endocrinol Metab. 34:666–681. 2023. View Article : Google Scholar : PubMed/NCBI

33 

Calejo I, Costa-Almeida R, Reis RL and Gomes ME: A physiology-inspired multifactorial toolbox in soft-to-hard musculoskeletal interface tissue engineering. Trends Biotechnol. 38:83–98. 2020. View Article : Google Scholar

34 

Adamičková A, Chomaničová N, Gažová A, Maďarič J, Červenák Z, Valášková S, Adamička M and Kyselovic J: Effect of atorvastatin on angiogenesis-related genes VEGF-A, HGF and IGF-1 and the modulation of PI3K/AKT/mTOR transcripts in bone-marrow-derived mesenchymal stem cells. Curr Issues Mol Biol. 45:2326–2337. 2023. View Article : Google Scholar

35 

Xu L, Zhao Q, Li K, Zhang Y, Wang C, Hind K, Wang L, Liu Y and Cheng X: The role of sex hormones on bone mineral density, marrow adiposity, and muscle adiposity in middle-aged and older men. Front Endocrinol (Lausanne). 13:8174182022. View Article : Google Scholar : PubMed/NCBI

36 

Wang Z, Wang Y, Tang Y, Guo X, Gao Q, Shao Y, Wang J, Tian R and Shi Y: Sodium benzoate inhibits osteoblast differentiation and accelerates bone loss by regulating the FGF2/p38/RUNX2 pathway. J Agric Food Chem. 73:13891–13901. 2025. View Article : Google Scholar : PubMed/NCBI

37 

Ogura H, Nakamura T, Ishii T, Saito A, Onodera S, Yamaguchi A, Nishii Y and Azuma T: Mechanical stress-induced FGF-2 promotes proliferation and consequently induces osteoblast differentiation in mesenchymal stem cells. Biochem Biophys Res Commun. 684:1491452023. View Article : Google Scholar : PubMed/NCBI

38 

Bakker AD and Jaspers RT: IL-6 and IGF-1 signaling within and between muscle and bone: How important is the mTOR pathway for bone metabolism? Curr Osteoporos Rep. 13:131–139. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Yoshida T and Delafontaine P: Mechanisms of IGF-1-mediated regulation of skeletal muscle hypertrophy and atrophy. Cells. 9:19702020. View Article : Google Scholar : PubMed/NCBI

40 

Chai J, Xu L and Liu N: miR-23b-3p regulates differentiation of osteoclasts by targeting PTEN via the PI3k/AKT pathway. Arch Med Sci. 18:1542–1557. 2019.PubMed/NCBI

41 

Liu X, Liu L, Chen K, Sun L, Li W and Zhang S: Huaier shows anti-cancer activities by inhibition of cell growth, migration and energy metabolism in lung cancer through PI3K/AKT/HIF-1α pathway. J Cell Mol Med. 25:2228–2237. 2021. View Article : Google Scholar

42 

Hang K, Wang Y, Bai J, Wang Z, Wu W, Zhu W, Liu S, Pan Z, Chen J and Chen W: Chaperone-mediated autophagy protects the bone formation from excessive inflammation through PI3K/AKT/GSK3β/β-catenin pathway. FASEB J. 38:e236462024. View Article : Google Scholar

43 

Peifer C, Oláh T, Venkatesan JK, Goebel L, Orth P, Schmitt G, Zurakowski D, Menger MD, Laschke MW, Cucchiarini M and Madry H: locally directed recombinant adeno-associated virus-mediated igf-1 gene therapy enhances osteochondral repair and counteracts early osteoarthritis in vivo. Am J Sports Med. 52:1336–1349. 2024. View Article : Google Scholar : PubMed/NCBI

44 

Zhang C, Wang J, Xie Y, Wang L, Yang L, Yu J, Miyamoto A and Sun F: Development of FGF-2-loaded electrospun waterborne polyurethane fibrous membranes for bone regeneration. Regen Biomater. 8:rbaa0462020. View Article : Google Scholar

45 

Kogure K, Hasuike A, Kurachi R, Igarashi Y, Idesawa M and Sato S: Effect of a recombinant human basic fibroblast growth factor 2 (rhFGF-2)-Impregnated atelocollagen sponge on vertical guided bone regeneration in a rat calvarial model. Dent J (Basel). 13:1772025. View Article : Google Scholar : PubMed/NCBI

46 

Yu X, Qi Y, Zhao T, Fang J, Liu X, Xu T, Yang Q and Dai X: NGF increases FGF2 expression and promotes endothelial cell migration and tube formation through PI3K/Akt and ERK/MAPK pathways in human chondrocytes. Osteoarthritis Cartilage. 27:526–534. 2019. View Article : Google Scholar

47 

Yin J, Qiu S, Shi B, Xu X, Zhao Y, Gao J, Zhao S and Min S: Controlled release of FGF-2 and BMP-2 in tissue engineered periosteum promotes bone repair in rats. Biomed Mater. 13:0250012018. View Article : Google Scholar : PubMed/NCBI

48 

Wu H, Yin G, Pu X, Wang J, Liao X and Huang Z: Inhibitory effects of combined bone morphogenetic protein 2, vascular endothelial growth factor, and basic fibroblast growth factor on osteoclast differentiation and activity. Tissue Eng Part A. 27:1387–1398. 2021. View Article : Google Scholar : PubMed/NCBI

49 

Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, Gho YS, Kurochkin IV, Mathivanan S, Quesenberry P, et al: Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 3:269132014. View Article : Google Scholar : PubMed/NCBI

50 

Akbar N, Paget D and Choudhury RP: Extracellular vesicles in innate immune cell programming. Biomedicines. 9:7132021. View Article : Google Scholar : PubMed/NCBI

51 

Garzetti L, Menon R, Finardi A, Bergami A, Sica A, Martino G, Comi G, Verderio C, Farina C and Furlan R: Activated macrophages release microvesicles containing polarized M1 or M2 mRNAs. J Leukoc Biol. 95:817–825. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Guan X, Li C, Wang X, Yang L, Lu Y and Guo Z: Engineered M2 macrophage-derived exosomes: mechanisms and therapeutic potential in inflammation regulation and regenerative medicine. Acta Biomater. 203:38–58. 2025. View Article : Google Scholar : PubMed/NCBI

53 

Mäki-Mantila K, Niskanen EA, Kainulainen K, Pardas LP, Aaltonen N, Wahbi W, Takabe P, Rönkä A, Rilla K and Pasonen-Seppänen S: Extracellular vesicles derived from pro-inflammatory M1 macrophages induce an inflammatory and invasive phenotype in melanoma cells. Cell Commun Signal. 24:102025. View Article : Google Scholar : PubMed/NCBI

54 

Vizoso FJ, Eiro N, Cid S, Schneider J and Perez-Fernandez R: Mesenchymal stem cell secretome: Toward cell-free therapeutic strategies in regenerative medicine. Int J Mol Sci. 18:18522017. View Article : Google Scholar : PubMed/NCBI

55 

Liu S, Chen J, Shi J, Zhou W, Wang L, Fang W, Zhong Y, Chen X, Chen Y, Sabri A and Liu S: M1-like macrophage-derived exosomes suppress angiogenesis and exacerbate cardiac dysfunction in a myocardial infarction microenvironment. Basic Res Cardiol. 115:222020. View Article : Google Scholar : PubMed/NCBI

56 

Li Z, Wang Y, Li S and Li Y: Exosomes derived from M2 macrophages facilitate osteogenesis and reduce adipogenesis of BMSCs. Front Endocrinol (Lausanne). 12:6803282021. View Article : Google Scholar : PubMed/NCBI

57 

Zhang Y, Liang Y and Zhou Y: M2 polarization of RAW264.7-derived exosomes inhibits osteoclast differentiation and inflammation via PKM2/HIF-1α axis. Immunol Invest. 54:1195–1209. 2025. View Article : Google Scholar : PubMed/NCBI

58 

Huang R, Wang X, Zhou Y and Xiao Y: RANKL-induced M1 macrophages are involved in bone formation. Bone Res. 5:170192017. View Article : Google Scholar : PubMed/NCBI

59 

Kang M, Huang CC, Lu Y, Shirazi S, Gajendrareddy P, Ravindran S and Cooper LF: Bone regeneration is mediated by macrophage extracellular vesicles. Bone. 141:1156272020. View Article : Google Scholar : PubMed/NCBI

60 

Ge X, Tang P, Rong Y, Jiang D, Lu X, Ji C, Wang J, Huang C, Duan A, Liu Y, et al: Exosomal miR-155 from M1-polarized macrophages promotes Endo MT and impairs mitochondrial function via activating NF-κB signaling pathway in vascular endothelial cells after traumatic spinal cord injury. Redox Biol. 41:1019322021. View Article : Google Scholar

61 

Hu Y, Wang Y, Chen T, Hao Z, Cai L and Li J: Exosome: Function and application in inflammatory bone diseases. Oxid Med Cell Longev. 2021:63249122021. View Article : Google Scholar : PubMed/NCBI

62 

Pajarinen J, Lin T, Gibon E, Kohno Y, Maruyama M, Nathan K, Lu L, Yao Z and Goodman SB: Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials. 196:80–89. 2019. View Article : Google Scholar

63 

Paschalidi P, Gkouveris I, Soundia A, Kalfarentzos E, Vardas E, Georgaki M, Kostakis G, Erovic BM, Tetradis S, Perisanidis C and Nikitakis NG: The role of M1 and M2 macrophage polarization in progression of medication-related osteonecrosis of the jaw. Clin Oral Investig. 25:2845–2857. 2021. View Article : Google Scholar :

64 

Boldin MP and Baltimore D: MicroRNAs, new effectors and regulators of NF-κB. Immunol Rev. 246:205–220. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Salhotra A, Shah HN, Levi B and Longaker MT: Mechanisms of bone development and repair. Nat Rev Mol Cell Biol. 21:696–711. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Wang Z, Zhu H, Shi H, Zhao H, Gao R, Weng X, Liu R, Li X, Zou Y, Hu K, et al: Exosomes derived from M1 macrophages aggravate neointimal hyperplasia following carotid artery injuries in mice through miR-222/CDKN1B/CDKN1C pathway. Cell Death Dis. 10:4222019. View Article : Google Scholar : PubMed/NCBI

67 

Qi Y, Zhu T, Zhang T, Wang X, Li W, Chen D, Meng H and An S: M1 macrophage-derived exosomes transfer miR-222 to induce bone marrow mesenchymal stem cell apoptosis. Lab Invest. 101:1318–1326. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Li L, Zheng B, Zhang F, Luo X, Li F, Xu T, Zhao H, Shi G, Guo Y, Shi J and Sun J: LINC00370 modulates miR-222-3p-RGS4 axis to protect against osteoporosis progression. Arch Gerontol Geriatr. 97:1045052021. View Article : Google Scholar : PubMed/NCBI

69 

Jiang C, Xia W, Wu T, Pan C, Shan H, Wang F, Zhou Z and Yu X: Inhibition of microRNA-222 up-regulates TIMP3 to promotes osteogenic differentiation of MSCs from fracture rats with type 2 diabetes mellitus. J Cell Mol Med. 24:686–694. 2020. View Article : Google Scholar

70 

Yu L, Hu M, Cui X, Bao D, Luo Z, Li D, Li L, Liu N, Wu Y, Luo X and Ma Y: M1 macrophage-derived exosomes aggravate bone loss in postmenopausal osteoporosis via a microRNA-98/DUSP1/JNK axis. Cell Biol Int. 45:2452–2463. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Peng S, Yan Y, Li R, Dai H and Xu J: Extracellular vesicles from M1-polarized macrophages promote inflammation in the temporomandibular joint via miR-1246 activation of the Wnt/β-catenin pathway. Ann N Y Acad Sci. 1503:48–59. 2021. View Article : Google Scholar : PubMed/NCBI

72 

He XT, Li X, Yin Y, Wu RX, Xu XY and Chen FM: The effects of conditioned media generated by polarized macrophages on the cellular behaviours of bone marrow mesenchymal stem cells. J Cell Mol Med. 22:1302–1315. 2018. View Article : Google Scholar :

73 

Xia Y, He XT, Xu XY, Tian BM, An Y and Chen FM: Exosomes derived from M0, M1 and M2 macrophages exert distinct influences on the proliferation and differentiation of mesenchymal stem cells. PeerJ. 8:e89702020. View Article : Google Scholar : PubMed/NCBI

74 

Schlundt C, Fischer H, Bucher CH, Rendenbach C, Duda GN and Schmidt-Bleek K: The multifaceted roles of macrophages in bone regeneration: A story of polarization, activation and time. Acta Biomater. 133:46–57. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Wei F, Zhou Y, Wang J, Liu C and Xiao Y: The immunomodulatory role of BMP-2 on macrophages to accelerate osteogenesis. Tissue Eng Part A. 24:584–594. 2018. View Article : Google Scholar

76 

Wang J, Xue Y, Wang Y, Liu C, Hu S, Zhao H, Gu Q, Yang H, Huang L, Zhou X and Shi Q: BMP-2 functional polypeptides relieve osteolysis via bi-regulating bone formation and resorption coupled with macrophage polarization. NPJ Regen Med. 8:62023. View Article : Google Scholar : PubMed/NCBI

77 

Bouchareychas L, Duong P, Covarrubias S, Alsop E, Phu TA, Chung A, Gomes M, Wong D, Meechoovet B, Capili A, et al: Macrophage exosomes resolve atherosclerosis by regulating hematopoiesis and inflammation via microRNA cargo. Cell Rep. 32:1078812020. View Article : Google Scholar : PubMed/NCBI

78 

Yu S, Geng Q, Pan Q, Liu Z, Ding S, Xiang Q, Sun F, Wang C, Huang Y and Hong A: miR-690, a Runx2-targeted miRNA, regulates osteogenic differentiation of C2C12 myogenic progenitor cells by targeting NF-kappaB p65. Cell Biosci. 6:102016. View Article : Google Scholar : PubMed/NCBI

79 

Yang Y, Guo Z, Chen W, Wang X, Cao M, Han X, Zhang K, Teng B, Cao J, Wu W, et al: M2 macrophage-derived exosomes promote angiogenesis and growth of pancreatic ductal adenocarcinoma by targeting E2F2. Mol Ther. 29:1226–1238. 2021. View Article : Google Scholar :

80 

Xu T, Luo Y, Wang J, Zhang N, Gu C, Li L, Qian D, Cai W, Fan J and Yin G: Exosomal miRNA128-3pfrom mesenchymal stem cells of aged rats regulates osteogenesis and bone fracture healing by targeting Smad5. J Nanobiotechnology. 18:472020. View Article : Google Scholar

81 

Zhang D, Wu Y, Li Z, Chen H, Huang S, Jian C and Yu A: miR-144-5p, an exosomal miRNA from bone marrow-derived macrophage in type 2 diabetes, impairs bone fracture healing via targeting Smad1. J Nanobiotechnology. 19:2262021. View Article : Google Scholar : PubMed/NCBI

82 

Huang X, Xiong X, Liu J, Zhao Z and Cen X: MicroRNAs-containing extracellular vesicles in bone remodeling: anemerging frontier. Life Sci. 254:1178092020. View Article : Google Scholar

83 

Luo ML, Jiao Y, Gong WP, Li Y, Niu LN, Tay FR and Chen JH: Macrophages enhance mesenchymal stem cell osteogenesis via down-regulation of reactive oxygen species. J Dent. 94:1032972020. View Article : Google Scholar : PubMed/NCBI

84 

Ren Q, Xing W, Jiang B, Feng H, Hu X, Suo J, Wang L and Zou W: Tenascin-C promotes bone regeneration via inflammatory macrophages. Cell Death Differ. 32:763–775. 2025. View Article : Google Scholar : PubMed/NCBI

85 

Halper J, Dolfi B, Ivanov S, Madel MB and Blin-Wakkach C: Macrophages and osteoclasts: Similarity and divergence between bone phagocytes. Front Immunol. 16:16838722025. View Article : Google Scholar : PubMed/NCBI

86 

Hao W, Chen S, Chao H, Li Z, Yang H, Chen D, Li S, Zhang S, Zhang J, Wang J, et al: IL-33-Induced TREM2(+) macrophages promote pathological new bone formation through CREG1-IGF2R axis in ankylosing spondylitis. Adv Sci (Weinh). 12:e25009522025. View Article : Google Scholar : PubMed/NCBI

87 

Choi JS, Yoon HI, Lee KS, Choi YC, Yang SH, Kim IS and Cho YW: Exosomes from differentiating human skeletal muscle cells trigger myogenesis of stem cells and provide biochemical cues for skeletal muscle regeneration. J Control Release. 222:107–115. 2016. View Article : Google Scholar

88 

Forterre A, Jalabert A, Berger E, Baudet M, Chikh K, Errazuriz E, De Larichaudy J, Chanon S, Weiss-Gayet M, Hesse AM, et al: Proteomic analysis of C2C12 myoblast and myotube exosome-like vesicles: A new paradigm for myoblast-myotube cross talk? PLoS One. 9:e841532014. View Article : Google Scholar : PubMed/NCBI

89 

Mobley CB, Mumford PW, McCarthy JJ, Miller ME, Young KC, Martin JS, Beck DT, Lockwood CM and Roberts MD: Whey protein-derived exosomes increase protein synthesis and hypertrophy in C2-C12 myotubes. J Dairy Sci. 100:48–64. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Yin J, Qian Z, Chen Y, Li Y and Zhou X: MicroRNA regulatory networks in the pathogenesis of sarcopenia. J Cell Mol Med. 24:4900–4912. 2020. View Article : Google Scholar : PubMed/NCBI

91 

Gopal Krishnan PD, Lee WX, Goh KY, Choy SM, Turqueza LRR, Lim ZH and Tang HW: Transcriptional regulation of autophagy in skeletal muscle stem cells. Dis Model Mech. 18:DMM0520072025. View Article : Google Scholar : PubMed/NCBI

92 

Chen W, Chen Y, Liu Y and Wang X: Autophagy in muscle regeneration: Potential therapies for myopathies. J Cachexia Sarcopenia Muscle. 13:1673–1685. 2022. View Article : Google Scholar : PubMed/NCBI

93 

Guescini M, Canonico B, Lucertini F, Maggio S, Annibalini G, Barbieri E, Luchetti F, Papa S and Stocchi V: Muscle releases alpha-sarcoglycan positive extracellular vesicles carrying miRNAs in the bloodstream. PLoS One. 10:e01250942015. View Article : Google Scholar : PubMed/NCBI

94 

Chaturvedi P, Kalani A, Medina I, Familtseva A and Tyagi SC: Cardiosome mediated regulation of MMP9 in diabetic heart: Role of mir29b and mir455 in exercise. J Cell Mol Med. 19:2153–2161. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020. View Article : Google Scholar : PubMed/NCBI

96 

Qin W and Dallas SL: Exosomes and extracellular RNA in muscle and bone aging and crosstalk. Curr Osteoporos Rep. 17:548–559. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Herrmann M, Engelke K, Ebert R, Müller-Deubert S, Rudert M, Ziouti F, Jundt F, Felsenberg D and Jakob F: Interactions between muscle and bone-where physics meets biology. Biomolecules. 10:4322020. View Article : Google Scholar : PubMed/NCBI

98 

Li G, Zhang L, Wang D, AIQudsy L, Jiang JX, Xu H and Shang P: Muscle-bone crosstalk and potential therapies for sarco-osteoporosis. J Cell Biochem. 120:14262–14273. 2019. View Article : Google Scholar : PubMed/NCBI

99 

Xu Q, Cui Y, Luan J, Zhou X, Li H and Han J: Exosomes from C2C12 myoblasts enhance osteogenic differentiation of MC3T3-E1 pre-osteoblasts by delivering miR-27a-3p. Biochem Biophys Res Commun. 498:32–37. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Kitase Y, Vallejo JA, Gutheil W, Vemula H, Jähn K, Yi J, Zhou J, Brotto M and Bonewald LF: β-aminoiso-butyric acid, 1-BAIBA, is a muscle-derived osteocyte survival factor. Cell Rep. 22:1531–1544. 2018. View Article : Google Scholar : PubMed/NCBI

101 

Li Y, Wang X, Pan C, Yuan H, Li X, Chen Z and He H: Myoblast-derived exosomal Prrx2 attenuates osteoporosis via transcriptional regulation of lncRNA-MIR22HG to activate Hippo pathway. Mol Med. 29:542023. View Article : Google Scholar : PubMed/NCBI

102 

Li H, Lin X, Yang D, Chen Z, Wang X, Re F, Wei J and Chen J: Cancer-associated fibroblasts support bone tropic metastasis by acting as coordinators between the tumor microenvironment and bone matrix in breast cancer. Neoplasma. 68:10–22. 2021. View Article : Google Scholar

103 

Zheng YL, Song G, Guo JB, Su X, Chen YM, Yang Z, Chen PJ and Wang XQ: Interactions among lncRNA/circRNA, miRNA, and mRNA in musculoskeletal degenerative diseases. Front Cell Dev Biol. 9:7539312021. View Article : Google Scholar : PubMed/NCBI

104 

Toita R, Shimizu Y, Shimizu E, Deguchi T, Tsuchiya A, Kang JH, Kitamura M, Kato A, Yamada H, Yamaguchi S and Kasahara S: Collagen patches releasing phosphatidylserine liposomes guide M1-to-M2 macrophage polarization and accelerate simultaneous bone and muscle healing. Acta Biomater. 187:51–65. 2024. View Article : Google Scholar : PubMed/NCBI

105 

Wehbe Z, Wehbe M, Al Khatib A, Dakroub AH, Pintus G, Kobeissy F and Eid AH: Emerging understandings of the role of exosomes in atherosclerosis. J Cell Physiol. 240:e314542025. View Article : Google Scholar :

106 

An F, Wang X, Wang C, Liu Y, Sun B, Zhang J, Gao P and Yan C: Research progress on the role of lncRNA-miRNA networks in regulating adipogenic and osteogenic differentiation of bone marrow mesenchymal stem cells in osteoporosis. Front Endocrinol (Lausanne). 14:12106272023. View Article : Google Scholar : PubMed/NCBI

107 

Liu K, Luo X, Lv ZY, Zhang YJ, Meng Z, Li J, Meng CX, Qiang HF, Hou CY, Hou L, et al: Macrophage-derived exosomes promote bone mesenchymal stem cells towards osteoblastic fate through microRNA-21a-5p. Front Bioeng Biotechnol. 9:8014322022. View Article : Google Scholar : PubMed/NCBI

108 

Xiong Y, Tang Y, Fan F, Zeng Y, Li C, Zhou G, Hu Z, Zhang L and Liu Z: Exosomal hsa-miR-21-5p derived from growth hormone-secreting pituitary adenoma promotes abnormal bone formation in acromegaly. Transl Res. 215:1–16. 2020. View Article : Google Scholar

109 

Choi SH, Chung KY, Johnson BJ, Go GW, Kim KH, Choi CW and Smith SB: Co-culture of bovine muscle satellite cells with preadipocytes increases PPARү and C/EBPβ gene expression in differentiated myoblasts and increases GPR43 gene expression in adipocytes. J Nutr Biochem. 24:539–543. 2013. View Article : Google Scholar

110 

Yuan R and Li J: Role of macrophages andtheir exosomes in orthopedic diseases. Peer J. 12:e171462024.

111 

Li K, Yan G, Huang H, Zheng M, Ma K, Cui X, Lu D, Zheng L, Zhu B, Cheng J and Zhao J: Anti-inflammatory and immunomodulatory effects of the extracellular vesicles derived from human umbilical cord mesenchymal stem cells on osteoarthritis via M2 macrophages. J Nanobiotechnology. 20:382022. View Article : Google Scholar : PubMed/NCBI

112 

Pu P, Wu S, Zhang K, Xu H, Guan J, Jin Z, Sun W, Zhang H and Yan B: Mechanical force induces macrophage-derived exosomal UCHL3 promoting bone marrow mesenchymal stem cell osteogenesis by targeting SMAD1. J Nanobiotechnology. 21:882023. View Article : Google Scholar : PubMed/NCBI

113 

Zhdanov VP: Interplay of cellular mRNA, miRNA and Viral miRNA during infection of a cell. Int J Mol Sci. 24:1222022. View Article : Google Scholar

114 

Bin-Bin Z, Da-Wa ZX, Chao L, Lan-Tao Z, Tao W, Chuan L, Chao-Zheng L, De-Chun L, Chang F, Shu-Qing W, et al: M2 macrophagy-derived exosomal miRNA-26a-5p induces osteogenic differentiation of bone mesenchymal stem cells. J Orthop Surg Res. 17:1372022. View Article : Google Scholar : PubMed/NCBI

115 

Qi L, Hong S, Zhao T, Yan J, Ge W, Wang J, Fang X, Jiang W, Shen SG and Zhang L: DNA Tetrahedron Delivering miR-21-5p promotes senescent bone defects repair through synergistic regulation of osteogenesis and angiogenesis. Adv Healthc Mater. 13:e24012752024. View Article : Google Scholar : PubMed/NCBI

116 

Hrdlicka HC, Pereira RC, Shin B, Yee SP, Deymier AC, Lee SK and Delany AM: Inhibition of miR-29-3p isoforms via tough decoy suppresses osteoblast function in homeostasis but promotes intermittent parathyroid hormone-induced bone anabolism. Bone. 143:1157792021. View Article : Google Scholar

117 

Gu Y, Ma L, Song L, Li X, Chen D and Bai X: miR-155 inhibits mouse Osteoblast differentiation by suppressing SMAD5 expression. Biomed Res Int. 2017:18935202017. View Article : Google Scholar : PubMed/NCBI

118 

Lan S and Albinsson S: Regulation of IRS-1, insulin signaling and glucose uptake by miR-143/145 in vascular smooth muscle cells. Biochem Biophys Res Commun. 529:119–125. 2020. View Article : Google Scholar : PubMed/NCBI

119 

Hade MD, Suire CN and Suo Z: Mesenchymal stem cell-derived exosomes: Applications in regenerative medicine. Cells. 10:19592021. View Article : Google Scholar : PubMed/NCBI

120 

Mezil Y, Obeid J, Raha S, Hawke TJ and Timmons BW: The Systemic Effects of Exercise on Regulators of Muscle and Bone in Girls and Women. Pediatr Exerc Sci. 32:117–123. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Zhang J, Rong Y, Luo C and Cui W: Bone marrow mesenchymal stem cell-derived exosomes prevent osteoarthritis by regulating synovial macrophage polarization. Aging (Albany NY). 12:25138–25152. 2020. View Article : Google Scholar : PubMed/NCBI

122 

Qin M, Zhu J, Xing L, Fan Y, Luo J, Sun J, Chen T, Zhang Y and Xi Q: Adipose-derived exosomes ameliorate skeletal muscle atrophy via miR-146a-5p/IGF-1R signaling. J Nanobiotechnology. 22:7542024. View Article : Google Scholar : PubMed/NCBI

123 

Hu GF, Wang C, Hu GX, Wu G, Zhang C, Zhu W, Chen C, Gu Y, Zhang H and Yang Z: AZD3463, an IGF-1R inhibitor, suppresses breast cancer metastasis to bone via modulation of the PI3K-Akt pathway. Ann Transl Med. 8:3362020. View Article : Google Scholar : PubMed/NCBI

124 

Cao D, Lei Y, Ye Z, Zhao L, Wang H, Zhang J, He F, Huang L, Shi D, Liu Q, et al: Blockade of IGF/IGF-1R signaling axis with soluble IGF-1R mutants suppresses the cell proliferation and tumor growth of human osteosarcoma. Am J Cancer Res. 10:3248–3266. 2020.PubMed/NCBI

125 

Song X, Xue Y, Fan S, Hao J and Deng R: Lipopolysaccharide-activated macrophages regulate the osteogenic differentiation of bone marrow mesenchymal stem cells through exosomes. PeerJ. 10:e134422022. View Article : Google Scholar : PubMed/NCBI

126 

Shu C, Smith SM, Little CB and Melrose J: Use of FGF-2 and FGF-18 to direct bone marrow stromal stem cells to chondrogenic and osteogenic lineages. Future Sci OA. 2:FSO1422016. View Article : Google Scholar

127 

Adhikary S, Choudhary D, Tripathi AK, Karvande A, Ahmad N, Kothari P and Trivedi R: FGF-2 targets sclerostin in bone and myostatin in skeletal muscle to mitigate the deleterious effects of glucocorticoid on musculoskeletal degradation. Life Sci. 229:261–276. 2019. View Article : Google Scholar : PubMed/NCBI

128 

Le Blanc S, Simann M, Jakob F, Schütze N and Schilling T: Fibroblast growth factors 1 and 2 inhibit adipogenesis of human bone marrow stromal cells in 3D collagen gels. Exp Cell Res. 338:136–148. 2015. View Article : Google Scholar : PubMed/NCBI

129 

Yang F, Chen C, Yang C, Chen R, Liu Z, Wen L, Xiao H, Zhou L, Geng B and Xia Y: Exosome-mediated perturbation of the immune-bone metabolism axis: A mechanistic investigation into bone loss in a simulated microgravity environment. Artif Cells Nanomed Biotechnol. 53:494–513. 2025. View Article : Google Scholar : PubMed/NCBI

130 

Hu K and Olsen BR: Osteoblast-derived VEGF regulates osteoblast differentiation and bone formation during bone repair. J Clin Invest. 126:509–526. 2016. View Article : Google Scholar : PubMed/NCBI

131 

Jiménez-Ortega RF, Ortega-Meléndez AI, Patiño N, Rivera-Paredez B, Hidalgo-Bravo A and Velázquez-Cruz R: The involvement of microRNAs in bone remodeling signaling pathways and their role in the development of osteoporosis. Biology (Basel). 13:5052024.PubMed/NCBI

132 

Geng Z, Sun T, Yu J, Wang N, Jiang Q, Wang P, Yang G, Li Y, Ding Y, Zhang J, et al: cinobufagin suppresses lipid peroxidation and inflammation in osteoporotic mice by promoting the delivery of miR-3102-5p by macrophage-derived exosomes. Int J Nanomedicine. 19:10497–10512. 2024. View Article : Google Scholar : PubMed/NCBI

133 

Hosseinpour S, Dai H, Walsh LJ and Xu C: Mesoporous core-cone silica nanoparticles can deliver mirna-26a to macrophages to exert immunomodulatory effects on osteogenesis in vitro. Nanomaterials (Basel). 13:17552023. View Article : Google Scholar : PubMed/NCBI

134 

Liu J, Sun Z, You Y, Zhang L, Hou D, Gu G, Chen Y and Jiao G: M2 macrophage-derived exosomal miR-486-5p influences the differentiation potential of bone marrow mesenchymal stem cells and osteoporosis. Aging (Albany NY). 15:9499–9520. 2023. View Article : Google Scholar : PubMed/NCBI

135 

Zhao W, Zhang S, Wang B, Huang J, Lu WW and Chen D: Runx2 and microRNA regulation in bone and cartilage diseases. Ann N Y Acad Sci. 1383:80–87. 2016. View Article : Google Scholar : PubMed/NCBI

136 

Faraldi M, Sansoni V and Lombardi G: Recent advances in the role of miRNAs in bone disease. Curr Opin Endocrinol Diabetes Obes. 32:149–155. 2025. View Article : Google Scholar : PubMed/NCBI

137 

Chen G, Deng C and Li YP: TGF-β and BMP signaling in osteoblast differentiation and bone formation. Int J Biol Sci. 8:272–288. 2012. View Article : Google Scholar :

138 

Kuroyanagi G, Tokuda H, Fujita K, Kawabata T, Sakai G, Kim W, Hioki T, Tachi J, Matsushima-Nishiwaki R, Otsuka T, et al: Upregulation of TGF-β-induced HSP27 by HSP90 inhibitors in osteoblasts. BMC Musculoskelet Disord. 23:4952022. View Article : Google Scholar

139 

Wu Y, Zhang C and Lv C: Direct and indirect regulation of bone metabolism by lactoferrin. Front Endocrinol (Lausanne). 16:16603122025. View Article : Google Scholar : PubMed/NCBI

140 

Méndez-Mancilla A, Turiján-Espinoza E, Vega-Cárdenas M, Hernández-Hernández GE, Uresti-Rivera EE, Vargas-Morales JM and Portales-Pérez DP: miR-21, miR-221, miR-29 and miR-34 are distinguishable molecular features of a metabolically unhealthy phenotype in young adults. PLoS One. 19:e03004202024. View Article : Google Scholar : PubMed/NCBI

141 

Fu X, Li Y, Huang T, Yu Z, Ma K, Yang M, Liu Q, Pan H, Wang H, Wang J and Guan M: Runx2/Osterix and zinc uptake synergize to orchestrate osteogenic differentiation and citrate containing bone apatite formation. Adv Sci (Weinh). 5:17007552018.PubMed/NCBI

142 

Luo K: Signaling cross talk between TGF-β/Smad and other signaling pathways. Cold Spring Harb Perspect Biol. 9:a0221372017.

143 

Zou ML, Chen ZH, Teng YY, Liu SY, Jia Y, Zhang KW, Sun ZL, Wu JJ, Yuan ZD, Feng Y, et al: The Smad Dependent TGF-beta and BMP signaling pathway in bone remodeling and therapies. Front Mol Biosci. 8:5933102021.

144 

Zhang B, Zhang X, Xiao J, Zhou X, Chen Y and Gao C: Neuropeptide Y upregulates Runx2 and osterix and enhances osteogenesis in mouse MC3T3-E1 cells via an autocrine mechanism. Mol Med Rep. 22:4376–4382. 2020.PubMed/NCBI

145 

Chen M, Li Y, Yang Z, Chen M, Li J, Bao G, Ma L and Hu J: M2-exo promote orthodontic bone remodeling via the MeCP2-TCF20-HDAC1 axis. Stem Cell Res Ther. 16:5692025.PubMed/NCBI

146 

Fu XH, Li JP, Li XY, Tan Y, Zhao M, Zhang SF, Wu XD and Xu JG: M2-Macrophage-Derived exosomes promote meningioma progression through TGF-β signaling pathway. J Immunol Res. 2022:83265912022.

147 

Li H, Yang Y, Gao Y, Li B, Yang J, Liu P, Zhang M and Ning G: Exosomes derived from hypoxia-preconditioned M2 macrophages alleviate degeneration in knee osteoarthritis through the miR-124-3p/STAT3 axis. J Transl Med. 23:7722025.

148 

Xie Q, Wei W, Ruan J, Ding Y, Zhuang A, Bi X, Sun H, Gu P, Wang Z and Fan X: Effects of miR-146a on the osteogenesis of adipose-derived mesenchymal stem cells and bone regeneration. Sci Rep. 7:428402017.PubMed/NCBI

149 

Ding S, Ma Y, Yang J, Tang Y, Jin Y, Li L and Ma C: MiR-224-5p inhibits osteoblast differentiation and impairs bone formation by targeting Runx2 and Sp7. Cytotechnology. 75:505–516. 2023.PubMed/NCBI

150 

Liao X, Yang Z, Li Y, Cui Y, Ma L, Liang C, Guan Z and Hu J: M2 macrophage-derived exosome facilitates aerobic glycolysis and osteogenic differentiation of hPDLSCs by regulating TRIM26-induced PKM ubiquitination. Free Radic Biol Med. 237:88–100. 2025.PubMed/NCBI

151 

Le G, Wen R, Fang H, Huang Z, Wang Y and Luo H: Exosomal miR-122 derived from M2 macrophages induces osteogenic differentiation of bone marrow mesenchymal stem cells in the treatment of alcoholic osteonecrosis of the femoral head. J Orthop Surg Res. 20:1072025. View Article : Google Scholar : PubMed/NCBI

152 

Xu Y, Jin Y, Hong F, Ma Y, Yang J, Tang Y, Zhu Z, Wu J, Bao Q, Li L, et al: MiR-664-3p suppresses osteoblast differentiation and impairs bone formation via targeting Smad4 and Osterix. J Cell Mol Med. 25:5025–5037. 2021. View Article : Google Scholar : PubMed/NCBI

153 

Sun WL, Wang N and Xu Y: Impact of miR-302b on calcium-phosphorus metabolism and vascular calcification of rats with chronic renal failure by regulating BMP-2/Runx2/Osterix signaling pathway. Arch Med Res. 49:164–171. 2018. View Article : Google Scholar : PubMed/NCBI

154 

Ling F, Bai J, Xie J, Liu J, Lu Q, Yuan L, Li H and Qian Z: Biomimetic periosteum combining BMP-2-loaded M2 macrophage-derived exosomes for enhanced bone defect repair. Front Bioeng Biotechnol. 13:16393942025. View Article : Google Scholar : PubMed/NCBI

155 

Yan CP, Wang XK, Jiang K, Yin C, Xiang C, Wang Y, Pu C, Chen L and Li YL: β-Ecdysterone enhanced bone regeneration through the BMP-2/SMAD/RUNX2/Osterix signaling pathway. Front Cell Dev Biol. 10:8832282022. View Article : Google Scholar

156 

Zhang Y, Yu T, Xiang Q, van den Tillaart F, Ma J, Zhuang Z, Stessuk T, Wang H and van den Beucken JJJP: Osteoclasts drive bone formation in ectopic and orthotopic environments. Biomaterials. 322:1233772025. View Article : Google Scholar : PubMed/NCBI

157 

Chen X, Wan Z, Yang L, Song S, Fu Z, Tang K, Chen L and Song Y: Exosomes derived from reparative M2-like macrophages prevent bone loss in murine periodontitis models via IL-10 mRNA. J Nanobiotechnology. 20:1102022. View Article : Google Scholar : PubMed/NCBI

158 

Udagawa N, Koide M, Nakamura M, Nakamichi Y, Yamashita T, Uehara S, Kobayashi Y, Furuya Y, Yasuda H, Fukuda C and Tsuda E: Osteoclast differentiation by RANKL and OPG signaling pathways. J Bone Miner Metab. 39:19–26. 2021. View Article : Google Scholar

159 

Kim HJ, Kang WY, Seong SJ, Kim SY, Lim MS and Yoon YR: Follistatin-like 1 promotes osteoclast formation via RANKL-mediated NF-ĸB activation and M-CSF-induced precursor proliferation. Cell Signal. 28:1137–1144. 2016. View Article : Google Scholar : PubMed/NCBI

160 

Nakao Y, Fukuda T, Zhang Q, Sanui T, Shinjo T, Kou X, Chen C, Liu D, Watanabe Y, Hayashi C, et al: Exosomes from TNF-alpha-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater. 122:306–324. 2021. View Article : Google Scholar

161 

Chen S, Liu J and Zhu L: M2-like macrophage-derived exosomes inhibit osteoclastogenesis via releasing miR-1227-5p. Immunobiology. 230:1528612025. View Article : Google Scholar

162 

Takeda T, Tsubaki M, Genno S, Tomita K and Nishida S: RANK/RANKL axis promotes migration, invasion, and metastasis of osteosarcoma via activating NF-ĸB pathway. Exp Cell Res. 436:1139782024. View Article : Google Scholar

163 

Guo W, Li H, Lou Y, Zhang Y, Wang J, Qian M, Wei H, Xiao J and Xu Y: Tyloxapol inhibits RANKL-stimulated osteoclastogenesis and ovariectomized-induced bone loss by restraining NF-ĸB and MAPK activation. J OrthopTranslat. 28:148–158. 2021.

164 

Fan YS, Li Q, Hamdan N, Bian YF, Zhuang S, Fan K and Liu ZJ: Tetrahydroxystilbene glucoside regulates proliferation, differentiation, and OPG/RANKL/M-CSF expression in MC3T3-E1 Cells via the PI3K/Akt pathway. Molecules. 23:23062018. View Article : Google Scholar : PubMed/NCBI

165 

Drissi H and Sanjay A: The multifaceted osteoclast; far and beyond bone resorption. J Cell Biochem. 117:1753–1756. 2016. View Article : Google Scholar : PubMed/NCBI

166 

Guo ZY, Yin NN, Li XF, Wang MM, Sui XN, Jiang CD, Xu MH, Jia XE, Fu CJ, Chen TL and Liu X: Exosomes secreted from M2-polarized macrophages inhibit osteoclast differentiation via CYLD. Tissue Cell. 93:1026452025. View Article : Google Scholar

167 

Yasuda H: Discovery of the RANKL/RANK/OPG system. J Bone Miner Metab. 39:2–11. 2021. View Article : Google Scholar : PubMed/NCBI

168 

Hakki SS, Batoon L, Koh AJ, Kannan R, Mendoza-Reinoso V, Rubin J, Mccauley LK and Roca H: The effects ofpreosteoblast-derived exosomes on macrophages and bone in mice. J Cell Mol Med. 28:e180292024. View Article : Google Scholar

169 

Gebraad A, Kornilov R, Kaur S, Miettinen S, Haimi S, Peltoniemi H, Mannerström B and Seppänen-Kaijansinkko R: Monocyte-derived extracellular vesicles stimulate cytokine secretion and gene expression of matrix metalloproteinases by mesenchymal stem/stromal cells. FEBS J. 285:2337–2359. 2018. View Article : Google Scholar : PubMed/NCBI

170 

Zhu S, Yao F, Qiu H, Zhang G, Xu H and Xu J: Coupling factors and exosomal packaging microRNAs involved in the regulation of bone remodelling. Biol Rev Camb Philos Soc. 93:469–480. 2018. View Article : Google Scholar

171 

Liu S, Yan X, Guo J, An H, Li X, Yang L, Yu X and Li S: Periodontal ligament-associated protein-1 knockout mice regulate the differentiation of osteoclasts and osteoblasts through TGF-β1/Smad signaling pathway. J Cell Physiol. 239:e310622024. View Article : Google Scholar

172 

Stevenson J; medical advisory council of the British Menopause Society: Prevention and treatment of osteoporosis in women. Post Reprod Health. 29:11–14. 2023. View Article : Google Scholar :

173 

Händel MN, Cardoso I, von Bülow C, Rohde JF, Ussing A, Nielsen SM, Christensen R, Body JJ, Brandi ML, Diez-Perez A, et al: Fracture risk reduction and safety by osteoporosis treatment compared with placebo or active comparator in postmenopausal women: systematic review, network meta-analysis, and meta-regression analysis of randomised clinical trials. BMJ. 381:e0680332023. View Article : Google Scholar : PubMed/NCBI

174 

Tagliaferri C, Wittrant Y, Davicco MJ, Walrand S and Coxam V: Muscle and bone, two interconnected tissues. Ageing Res Rev. 21:55–70. 2015. View Article : Google Scholar : PubMed/NCBI

175 

Chen K, Jiao Y, Liu L, Huang M, He C, He W, Hou J, Yang M, Luo X and Li C: communications between bone marrow macrophages and bone cells in bone remodeling. Front Cell Dev Biol. 8:5982632020. View Article : Google Scholar

176 

Li Q, Gao H, Ma X, Wang Z, Zhao L and Xiao W: Exosome-mediated crosstalk between the cardiovascular and musculoskeletal systems: Mechanisms and therapeutic potential (Review). Int J Mol Med. 56:1292025. View Article : Google Scholar :

177 

Hou C, Zhang Y, Lv Z, Luan Y, Li J, Meng C, Liu K, Luo X, Chen L and Liu F: Macrophage exosomes modified by miR-365-2-5p promoted osteoblast osteogenic differentiation by targeting OLFML1. Regen Biomater. 11:rbae0182024. View Article : Google Scholar : PubMed/NCBI

178 

Hossain MA, Adithan A, Alam MJ, Kopalli SR, Kim B, Kang CW, Hwang KC and Kim JH: IGF-1 facilitates cartilage reconstruction by regulating PI3K/AKT, MAPK, and NF-kB signaling in rabbit osteoarthritis. J Inflamm Res. 14:3555–3568. 2021. View Article : Google Scholar : PubMed/NCBI

179 

Fuentes EN, Björnsson BT, Valdés JA, Einarsdottir IE, Lorca B, Alvarez M and Molina A: IGF-I/PI3K/Akt and IGF-I/MAPK/ERK pathways in vivo in skeletal muscle are regulated by nutrition and contribute to somatic growth in the fine flounder. Am J Physiol Regul Integr Comp Physiol. 300:R1532–R1542. 2011. View Article : Google Scholar : PubMed/NCBI

180 

Sudha S, Upmanyu A, Saraswat D and Singh M: Pharmacological impacts of tanshinone on osteogenesis and osteoclastogenesis: A review. Naunyn Schmiedebergs Arch Pharmacol. 398:135–146. 2025. View Article : Google Scholar

181 

Luo L, Avery SJ and Waddington RJ: Exploring a chemotactic role for EVs from progenitor cell populations of human exfoliated deciduous teeth for promoting migration of naïve BMSCs in bone repair process. Stem Cells Int. 2021:66817712021. View Article : Google Scholar

182 

Olivieri F, Prattichizzo F, Giuliani A, Matacchione G, Rippo MR, Sabbatinelli J and Bonafè M: miR-21 and miR-146a: The microRNAs of inflammaging and age-related diseases. Ageing Res Rev. 70:1013742021. View Article : Google Scholar : PubMed/NCBI

183 

Jann J, Gascon S, Roux S and Faucheux N: Influence of the TGF-β superfamily on osteoclasts/osteoblasts balance in physiological and pathological bone conditions. Int J Mol Sci. 21:75972020. View Article : Google Scholar

184 

Yu Y, Cai W, Xu Y and Zuo W: Down-regulation of miR-19b-3p enhances IGF-1 expression to induce osteoblast differentiation and improve osteoporosis. Cell Mol Biol (Noisy-le-grand). 68:160–168. 2022. View Article : Google Scholar : PubMed/NCBI

185 

Sabbieti MG, Agas D, Marchetti L, Coffin JD, Xiao L and Hurley MM: BMP-2 differentially modulates FGF-2 isoform effects in osteoblasts from newborn transgenic mice. Endocrinology. 154:2723–2733. 2013. View Article : Google Scholar : PubMed/NCBI

186 

Zhang C, Yang J, Zhu Z, Qin J, Yang L, Zhao X, Su W, Cai Y, Yang J, Wang F, et al: Exosomal lncRNA HOTAIR promotes osteoclast differentiation by targeting TGF-beta/PTHrP/RANKL pathway. Basic Clin Pharmacol Toxicol. 132:242–252. 2023. View Article : Google Scholar

187 

Wang J, Li X, Wang S, Cui J, Ren X and Su J: Bone-targeted exosomes: Strategies and applications. Adv Healthc Mater. 12:e22033612023. View Article : Google Scholar : PubMed/NCBI

188 

Sayer AA and Cruz-Jentoft A: Sarcopenia definition, diagnosis and treatment: consensus is growing. Age Ageing. 51:afac2202022. View Article : Google Scholar : PubMed/NCBI

189 

Jun L, Robinson M, Geetha T, Broderick TL and Babu JR: Prevalence and mechanisms of skeletal muscle atrophy in metabolic conditions. Int J Mol Sci. 24:29732023. View Article : Google Scholar : PubMed/NCBI

190 

Dong Q, Li D, Zhang K, Shi H, Cai M, Li Y, Zhao R and Qin D: Muscle-bone biochemical crosstalk in osteosarcopenia: Focusing on mechanisms and potential therapeutic strategies. J Endocrinol. 266:e2502342025. View Article : Google Scholar : PubMed/NCBI

191 

Zhang H, Du Y, Tang W, Chen M, Yu W, Ke Z, Dong S and Cheng Q: Eldecalcitol prevents muscle loss and osteoporosis in disuse muscle atrophy via NF-ĸB signaling in mice. Skelet Muscle. 13:222023. View Article : Google Scholar

192 

Bettis T, Kim BJ and Hamrick MW: Impact of muscle atrophy on bone metabolism and bone strength: Implications for muscle-bone crosstalk with aging and disuse. Osteoporos Int. 29:1713–1720. 2018. View Article : Google Scholar : PubMed/NCBI

193 

Rong S, Wang L, Peng Z, Liao Y, Li D, Yang X, Nuessler AK, Liu L, Bao W and Yang W: The mechanisms and treatments for sarcopenia: Could exosomes be a perspective research strategy in the future? J Cachexia Sarcopenia Muscle. 11:348–365. 2020. View Article : Google Scholar : PubMed/NCBI

194 

Chu R, Li M, Xie Y, Du Y and Ni T: Exercise interventions and serum IGF-1 levels in older adults with frailty and/or sarcopenia: A systematic review and meta analysis. Front Public Health. 13:16606942025. View Article : Google Scholar :

195 

Zhu J, Fan J, Xia Y, Wang H, Li Y, Feng Z and Fu C: Potential therapeutic targets of macrophages in inhibiting immune damage and fibrotic processes in musculoskeletal diseases. Front Immunol. 14:12194872023. View Article : Google Scholar : PubMed/NCBI

196 

Hu S, Wang S, Yang X, Li P, Li Z, Luo B, Liang Y and Pan X: Exosomes promise better bone regeneration. Regen Ther. 30:389–402. 2025. View Article : Google Scholar : PubMed/NCBI

197 

Liu W, Li L, Rong Y, Qian D, Chen J, Zhou Z, Luo Y, Jiang D, Cheng L, Zhao S, et al: Hypoxic mesenchymal stem cell-derived exosomes promote bone fracture healing by the transfer of miR-126. Acta Biomater. 103:196–212. 2020. View Article : Google Scholar

198 

Holliday LS, McHugh KP, Zuo J, Aguirre JI, Neubert JK and Rody WJ Jr: Exosomes: Novel regulators of bone remodelling and potential therapeutic agents for orthodontics. Orthod Craniofac Res. 20(Suppl 1): S95–S99. 2017. View Article : Google Scholar

199 

Hatakeyama J, Inoue S, Jiang H, Yokoi R and Moriyama H: Exercise-induced interactions between skeletal muscle and bone via myokines and osteokine in mice: Role of FNDC5/irisin, IGF-1, and osteocalcin. Bone. 190:1173142025. View Article : Google Scholar

200 

Krylova SV and Feng D: The machinery of exosomes: Biogenesis, release, and uptake. Int J Mol Sci. 24:13372023. View Article : Google Scholar : PubMed/NCBI

201 

Fridman E, Ginini L, Gil Z and Milman N: The purification and characterization of exosomes from macrophages. Methods Mol Biol. 2184:77–90. 2020. View Article : Google Scholar : PubMed/NCBI

202 

Xu WM, Li A, Chen JJ and Sun EJ: Research development on exosome separation technology. J Membr Biol. 256:25–34. 2023. View Article : Google Scholar

203 

Auquière M, Muccioli GG and des Rieux A: methods and challenges in purifying drug-loaded extracellular vesicles. J Extracell Vesicles. 14:e700972025. View Article : Google Scholar : PubMed/NCBI

204 

Yousaf I, Kegler U, Hofner M and Noehammer C: Evaluation of commercially available kits for parallel DNA and microRNA isolation suitable for epigenetic analyses from cell-free saliva and salivary extracellular vesicles. Int J Mol Sci. 26:63652025. View Article : Google Scholar : PubMed/NCBI

205 

Coughlan C, Bruce KD, Burgy O, Boyd TD, Michel CR, Garcia-Perez JE, Adame V, Anton P, Bettcher BM, Chial HJ, et al: Exosome isolation by ultracentrifugation and precipitation and techniques for downstream analyses. Curr Protoc Cell Biol. 88:e1102020. View Article : Google Scholar : PubMed/NCBI

206 

Sidhom K, Obi PO and Saleem A: Review of exosomal isolation methods: Is size exclusion chromatography the best option? Int J Mol Sci. 21:64662020. View Article : Google Scholar

207 

Gao M, Cai J, Zitkovsky HS, Chen B and Guo L: comparison of yield, purity, and functional properties of large-volume exosome isolation using ultrafiltration and polymer-based precipitation. Plast Reconstr Surg. 149:638–649. 2022. View Article : Google Scholar : PubMed/NCBI

208 

Greening DW, Xu R, Ji H, Tauro BJ and Simpson RJ: A protocol for exosome isolation and characterization: Evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol Biol. 1295:179–209. 2015. View Article : Google Scholar : PubMed/NCBI

209 

Contreras-Naranjo JC, Wu HJ and Ugaz VM: Microfluidics for exosome isolation and analysis: Enabling liquid biopsy for personalized medicine. Lab Chip. 17:3558–3577. 2017. View Article : Google Scholar : PubMed/NCBI

210 

Tang YT, Huang YY, Zheng L, Qin SH, Xu XP, An TX, Xu Y, Wu YS, Hu XM, Ping BH and Wang Q: Comparison of isolationmethods of exosomes and exosomal RNA from cell culture medium and serum. Int J Mol Med. 40:834–844. 2017. View Article : Google Scholar : PubMed/NCBI

211 

Huang Y, Liu Y, Huang Q, Sun S, Ji Z, Huang L, Li Z, Huang X, Deng W and Li T: TMT-Based quantitative proteomics analysis of synovial fluid-derived exosomes in inflammatory arthritis. Front Immunol. 13:8009022022. View Article : Google Scholar : PubMed/NCBI

212 

Huang LH, Rau CS, Wu SC, Wu YC, Wu CJ, Tsai CW, Lin CW, Lu TH and Hsieh CH: Identification and characterization of hADSC-derived exosome proteins from different isolation methods. J Cell Mol Med. 25:7436–7450. 2021. View Article : Google Scholar : PubMed/NCBI

213 

Lai JJ, Chau ZL, Chen SY, Hill JJ, Korpany KV, Liang NW, Lin LH, Lin YH, Liu JK, Liu YC, et al: Exosome processing and characterization approaches for research and technology development. Adv Sci (Weinh). 9:e21032222022. View Article : Google Scholar : PubMed/NCBI

214 

D'Acunzo P, Kim Y, Ungania JM, Pérez-González R, Goulbourne CN and Levy E: Isolation of mitochondria-derived mitovesicles and subpopulations of microvesicles and exosomes from brain tissues. Nat Protoc. 17:2517–2549. 2022. View Article : Google Scholar : PubMed/NCBI

215 

Doyle LM and Wang MZ: Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 8:7272019. View Article : Google Scholar : PubMed/NCBI

216 

Veerman RE, Teeuwen L, Czarnewski P, Güclüler Akpinar G, Sandberg A, Cao X, Pernemalm M, Orre LM, Gabrielsson S and Eldh M: Molecular evaluation of five different isolation methods for extracellular vesicles reveals different clinical applicability and subcellular origin. J Extracell Vesicles. 10:e121282021. View Article : Google Scholar : PubMed/NCBI

217 

Zhao R, Zhao T, He Z, Cai R and Pang W: Composition, isolation, identification and function of adipose tissue-derived exosomes. Adipocyte. 10:587–604. 2021. View Article : Google Scholar : PubMed/NCBI

218 

Corona ML, Hurbain I, Raposo G and van Niel G: Characterization of extracellular vesicles by transmission electron microscopy and immunolabeling electron microscopy. Methods Mol Biol. 2668:33–43. 2023. View Article : Google Scholar : PubMed/NCBI

219 

Wu X, Showiheen SAA, Sun AR, Crawford R, Xiao Y, Mao X and Prasadam I: Exosomes extraction and identification. Methods Mol Biol. 2054:81–91. 2019. View Article : Google Scholar : PubMed/NCBI

220 

Chen Z, Luo L, Ye T, Zhou J, Niu X, Yuan J, Yuan T, Fu D, Li H, Li Q and Wang Y: Identification of specific markers for human pluripotent stem cell-derived small extracellular vesicles. J Extracell Vesicles. 13:e124092024. View Article : Google Scholar : PubMed/NCBI

221 

Rahmatinejad F, Kharat Z, Jalili H, Renani MK and Mobasheri H: Comparison of morphology, protein concentration, and size distribution of bone marrow and Wharton's jelly-derived mesenchymal stem cells exosomes isolated by ultracentrifugation and polymer-based precipitation techniques. Tissue Cell. 88:1024272024. View Article : Google Scholar : PubMed/NCBI

222 

Kim J, Lyu HZ, Jung C, Lee KM, Han SH, Lee JH and Cha M: Osteogenic response of MC3T3-E1 and Raw264.7 in the 3D-encapsulated co-culture environment. Tissue Eng Regen Med. 18:387–397. 2021. View Article : Google Scholar : PubMed/NCBI

223 

Wragg NM, Mosqueira D, Blokpeol-Ferreras L, Capel A, Player DJ, Martin NRW, Liu Y and Lewis MP: Development of a 3D tissue-engineered skeletal muscle and bone co-culture system. Biotechnol J. 15:e19001062020. View Article : Google Scholar

224 

Ostrovidov S, Ahadian S, Ramon-Azcon J, Hosseini V, Fujie T, Parthiban SP, Shiku H, Matsue T, Kaji H, Ramalingam M, et al: Three-dimensional co-culture of C2C12/PC12 cells improves skeletal muscle tissue formation and function. J Tissue Eng Regen Med. 11:582–595. 2017. View Article : Google Scholar

225 

Liu J, Yang D, Shi S, Lin L, Xiao M, Yuan Z and Yu M: Overexpression of vasostatin-1 protects hypoxia/reoxygenation injuries in cardiomyocytes-endothelial cells transwell co-culture system. Cell Biol Int. 38:26–31. 2014. View Article : Google Scholar

226 

Liu M, Han Y, Wang J, Zhu Y, Zhang Y, Chu Q, Yang C, Chen B and Sun G: Skeletal muscle-derived IL-33 mediates muscle-to-bone crosstalk and regulates bone metabolism via CD8(+) T cell-secreted CCL5. EBioMedicine. 122:1060242025. View Article : Google Scholar : PubMed/NCBI

227 

Le VL, Chang CY, Chuang CW, Syu SH, Shih HJ, Nguyen Vo HP, Van MN and Huang CJ: Therapeutic effects of engineered exosomes from RAW264.7 Cells Overexpressing hsa-let-7i-5p against Sepsis in Mice-A comparative study with human placenta-derived mesenchymal stem cell exosomes. J Pers Med. 14:6192024. View Article : Google Scholar : PubMed/NCBI

228 

Bier A, Berenstein P, Kronfeld N, Morgoulis D, Ziv-Av A, Goldstein H, Kazimirsky G, Cazacu S, Meir R, Popovtzer R, et al: Placenta-derived mesenchymal stromal cells and their exosomes exert therapeutic effects in Duchenne muscular dystrophy. Biomaterials. 174:67–78. 2018. View Article : Google Scholar : PubMed/NCBI

229 

Cai H and Guo H: Mesenchymal stem cells and their exocytotic vesicles. Int J Mol Sci. 24:20852023. View Article : Google Scholar : PubMed/NCBI

230 

Zha Y, Li Y, Lin T, Chen J, Zhang S and Wang J: Progenitor cell-derived exosomes endowed with VEGF plasmids enhance osteogenic induction and vascular remodeling in large segmental bone defects. Theranostics. 11:397–409. 2021. View Article : Google Scholar : PubMed/NCBI

231 

Tenkumo T, Aobulikasimu A, Asou Y, Shirato M, Shishido S, Kanno T, Niwano Y, Sasaki K and Nakamura K: Proanthocyanidin-rich grape seed extract improves bone loss, bone healing, and implant osseointegration in ovariectomized animals. Sci Rep. 10:88122020. View Article : Google Scholar : PubMed/NCBI

232 

Halloran D, Pandit V, Chukwuocha K and Nohe A: Methyl-beta-cyclodextrin restores aberrant bone morphogenetic protein 2-signaling in bone marrow stromal cells obtained from aged C57BL/6 Mice. J Dev Biol. 12:302024. View Article : Google Scholar : PubMed/NCBI

233 

Yashima N, Minamizono W, Matsunaga H, Lyu J, Fujikawa K, Suito H, Okunuki T, Nakai S and Ohsako M: Non-contact electrical stimulation via a Vector-potential transformer promotes bone healing in drill-hole injury model. J Bone Miner Metab. 43:348–359. 2025. View Article : Google Scholar : PubMed/NCBI

234 

Muwanga GPB, Siliezar-Doyle J, Ortiz AA, Kaslow J, Haight ES and Tawfik VL: The tibial fracture-pin model: A clinically relevant mouse model of orthopedic injury. J Vis Exp. 28: View Article : Google Scholar : 2022.

235 

Wang BW, Jiang Y, Yao ZL, Chen PS, Yu B and Wang SN: Aucubin protects chondrocytes against IL-1β-Induced apoptosis in vitro and inhibits osteoarthritis in mice model. Drug Des Devel Ther. 13:3529–3538. 2019. View Article : Google Scholar :

236 

Suh HR, Cho HY and Han HC: Development of a novel model of intervertebral disc degeneration by the intradiscal application of monosodium iodoacetate (MIA) in rat. Spine J. 22:183–192. 2022. View Article : Google Scholar

237 

Li Y, Qiao X, Feng Y, Zhou R, Zhang K, Pan Y, Yan T, Yan L, Yang S, Wei X, et al: Characterization of the gut microbiota and fecal metabolome in the osteosarcoma mouse model. Aging (Albany NY). 16:10841–10859. 2024. View Article : Google Scholar : PubMed/NCBI

238 

Ferrena A, Wang J, Zhang R, Karadal-Ferrena B, Al-Hardan W, Singh S, Borjihan H, Schwartz EL, Zhao H, Oktay MH, et al: SKP2 knockout in Rb1/p53-Deficient mouse models of osteosarcoma induces immune infiltration and drives a transcriptional program with a favorable prognosis. Mol Cancer Ther. 23:223–234. 2024. View Article : Google Scholar :

239 

Wang C, Luo D, Zheng L and Zhao M: Anti-diabetic mechanism and potential bioactive peptides of casein hydrolysates in STZ/HFD-induced diabetic rats. J Sci Food Agric. 104:2947–2958. 2024. View Article : Google Scholar

240 

Yao Y, Cai X, Chen Y, Zhang M and Zheng C: Estrogen deficiency-mediated osteoimmunity in postmenopausal osteoporosis. Med Res Rev. 45:561–575. 2025. View Article : Google Scholar

241 

Rangel LBA, de Siqueira D, Soares ODR, Santana HS, Miguel EC, da Cunha M, Oliveira ALA, Pedrosa DF, Resgala LCR, Neto HAR, et al: Vitamin K supplementation modulates bone metabolism and ultra-structure of ovariectomized mice. Cell Physiol Biochem. 51:356–374. 2018. View Article : Google Scholar : PubMed/NCBI

242 

Chen MY, Zhao FL, Chu WL, Bai MR and Zhang DM: A review of tamoxifen administration regimen optimization for Cre/loxp system in mouse bone study. Biomed Pharmacother. 165:1150452023. View Article : Google Scholar : PubMed/NCBI

243 

Gelles K, Butylina M and Pietschmann P: Animal models for age-related osteoporosis. Gerontology. 71:1–29. 2025. View Article : Google Scholar : PubMed/NCBI

244 

Rong K, Chen P, Lang Y, Zhang Y, Wang Z, Wen F and Lu L: Morinda officinalis polysaccharide attenuates osteoporosis in rats underwent bilateral ovariectomy by suppressing the PGC-1α/PPARү pathway. J Orthop Surg (Hong Kong). 30:102255362211308242022. View Article : Google Scholar

245 

Liu Y, Dimango E, Bucovsky M, Agarwal S, Nishiyama K, Guo XE, Shane E and Stein EM: Abnormal microarchitecture and stiffness in postmenopausal women using chronic inhaled glucocorticoids. Osteoporos Int. 29:2121–2127. 2018. View Article : Google Scholar : PubMed/NCBI

246 

Abdi S, Javanmehr N, Ghasemi-Kasman M, Bali HY and Pirzadeh M: Stem cell-based therapeutic and diagnostic approaches in Alzheimer's disease. Curr Neuropharmacol. 20:1093–1115. 2022. View Article : Google Scholar : PubMed/NCBI

247 

Henderson S, Ibe I, Cahill S, Chung YH and Lee FY: Bone quality and fracture-healing in type-1 and type-2 diabetes mellitus. J Bone Joint Surg Am. 101:1399–1410. 2019. View Article : Google Scholar : PubMed/NCBI

248 

Castillo ÍMP, Argilés JM, Rueda R, Ramírez M and Pedrosa JML: Skeletal muscle atrophy and dysfunction in obesity and type-2 diabetes mellitus: Myocellular mechanisms involved. Rev Endocr Metab Disord. 26:815–836. 2025. View Article : Google Scholar : PubMed/NCBI

249 

Edgar L, Akbar N, Braithwaite AT, Krausgruber T, Gallart-Ayala H, Bailey J, Corbin AL, Khoyratty TE, Chai JT, Alkhalil M, et al: Hyperglycemia induces trained immunity in macrophages and their precursors and promotes atherosclerosis. Circulation. 144:961–982. 2021. View Article : Google Scholar : PubMed/NCBI

250 

Kishore A and Petrek M: roles of macrophage polarization and macrophage-derived miRNAs in pulmonary fibrosis. Front Immunol. 12:6784572021. View Article : Google Scholar : PubMed/NCBI

251 

Cazzanelli P, Lamoca M, Hasler J, Hausmann ON, Mesfin A, Puvanesarajah V, Hitzl W and Wuertz-Kozak K: The role of miR-155-5p in inflammation and mechanical loading during intervertebral disc degeneration. Cell Commun Signal. 22:4192024. View Article : Google Scholar : PubMed/NCBI

252 

Adamopoulos IE: Inflammation in bone physiology and pathology. Curr Opin Rheumatol. 30:59–64. 2018. View Article : Google Scholar :

253 

Sun Y, Kuek V, Liu Y, Tickner J, Yuan Y, Chen L, Zeng Z, Shao M, He W and Xu J: MiR-214 is an important regulator of the musculoskeletal metabolism and disease. J Cell Physiol. 234:231–245. 2018. View Article : Google Scholar : PubMed/NCBI

254 

Ma-On C, Sanpavat A, Whongsiri P, Suwannasin S, Hirankarn N, Tangkijvanich P and Boonla C: Oxidative stress indicated by elevated expression of Nrf2 and 8-OHdG promotes hepatocellular carcinoma progression. Med Oncol. 34:572017. View Article : Google Scholar : PubMed/NCBI

255 

Palma FR, Gantner BN, Sakiyama MJ, Kayzuka C, Shukla S, Lacchini R, Cunniff B and Bonini MG: ROS production by mitochondria: Function or dysfunction? Oncogene. 43:295–303. 2024. View Article : Google Scholar

256 

Song Y and Chung J: Aging aggravates periodontal inflammatory responses and alveolar bone resorption by porphyromonas gingivalis infection. Curr Issues Mol Biol. 45:6593–6604. 2023. View Article : Google Scholar : PubMed/NCBI

257 

Liu J, Zhao Y, Zhang Y, Yao X and Hang R: Exosomes derived from macrophages upon Zn ion stimulation promote osteoblast and endothelial cell functions. J Mater Chem B. 9:3800–3807. 2021. View Article : Google Scholar : PubMed/NCBI

258 

Zhu Y, Zhao S, Cheng L, Lin Z, Zeng M, Ruan Z, Sun B, Luo Z, Tang Y and Long H: Mg2+-mediated autophagy-dependent polarization of macrophages mediates the osteogenesis of bone marrow stromal stem cells by interfering with macrophage-derived exosomes containing miR-381. J Orthop Res. 40:1563–1576. 2022. View Article : Google Scholar

259 

Wei F, Li M, Crawford R, Zhou Y and Xiao Y: Exosome-integrated titanium oxide nanotubes for targeted bone regeneration. Acta Biomater. 86:480–492. 2019. View Article : Google Scholar : PubMed/NCBI

260 

Bai X, Gao M, Syed S, Zhuang J, Xu X and Zhang XQ: Bioactive hydrogels for bone regeneration. Bioact Mater. 3:401–417. 2018.PubMed/NCBI

261 

Moura MLA, Fugimoto M, Kawachi APM, de Oliveira ML, Lazaretti-Castro M and Reginato RD: Estrogen therapy associated with mechanical vibration improves bone microarchitecture and density in osteopenic female mice. J Anat. 233:715–723. 2018. View Article : Google Scholar : PubMed/NCBI

262 

Vilaca T, Eastell R and Schini M: Osteoporosis in men. Lancet Diabetes Endocrinol. 10:273–283. 2022. View Article : Google Scholar : PubMed/NCBI

263 

Cauley JA: Estrogen and bone health in men and women. Steroids. 99(Pt A): 11–15. 2015. View Article : Google Scholar : PubMed/NCBI

264 

Pellegrino A, Tiidus PM and Vandenboom R: Mechanisms of estrogen influence on skeletal muscle: Mass, regeneration, and mitochondrial function. Sports Med. 52:2853–2869. 2022. View Article : Google Scholar : PubMed/NCBI

265 

Alexander SE, Pollock AC and Lamon S: The effect of sex hormones on skeletal muscle adaptation in females. Eur J Sport Sci. 22:1035–1045. 2022. View Article : Google Scholar

266 

Jardí F, Laurent MR, Dubois V, Kim N, Khalil R, Decallonne B, Vanderschueren D and Claessens F: Androgen and estrogen actions on male physical activity: A story beyond muscle. J Endocrinol. 238:R31–R52. 2018. View Article : Google Scholar : PubMed/NCBI

267 

Unger CA, Aladhami AK, Hope MC, Cotham WE, Nettles KW, Clegg DJ, Velázquez KT and Enos RT: skeletal muscle endogenous estrogen production ameliorates the metabolic consequences of a high-fat diet in male mice. Endocrinology. 164:bqad1052023. View Article : Google Scholar : PubMed/NCBI

268 

Collins BC, Mader TL, Cabelka CA, Iñigo MR, Spangenburg EE and Lowe DA: Deletion of estrogen receptor α in skeletal muscle results in impaired contractility in female mice. J Appl Physiol (1985). 124:980–992. 2018. View Article : Google Scholar : PubMed/NCBI

269 

Tramunt B, Smati S, Grandgeorge N, Lenfant F, Arnal JF, Montagner A and Gourdy P: Sex differences in metabolic regulation and diabetes susceptibility. Diabetologia. 63:453–461. 2020. View Article : Google Scholar :

270 

O'Reilly J, Ono-Moore KD, Chintapalli SV, Rutkowsky JM, Tolentino T, Lloyd KCK, Olfert IM and Adams SH: Sex differences in skeletal muscle revealed through fiber type, capillarity, and transcriptomics profiling in mice. Physiol Rep. 9:e150312021.PubMed/NCBI

271 

Farhat F, Amérand A, Simon B, Guegueniat N and Moisan C: Gender-dependent differences of mitochondrial function and oxidative stress in rat skeletal muscle at rest and after exercise training. Redox Rep. 22:508–514. 2017. View Article : Google Scholar : PubMed/NCBI

272 

Clynes MA, Gregson CL, Bruyère O, Cooper C and Dennison EM: Osteosarcopenia: Where osteoporosis and sarcopenia collide. Rheumatology (Oxford). 60:529–537. 2021. View Article : Google Scholar

273 

Gálvez I, Navarro MC, Martín-Cordero L, Otero E, Hinchado MD and Ortega E: The influence of obesity and weight loss on the bioregulation of innate/inflammatory responses: Macrophages and immunometabolism. Nutrients. 14:6122022. View Article : Google Scholar : PubMed/NCBI

274 

Jin Z, Huang Q, Peng J, Liu Z, Hu R, Wu J and Wang F: MiR-125a-3p alleviates hyperproliferation of keratinocytes and psoriasis-like inflammation by targeting TLR4/NF-ĸB pathway. Postepy Dermatol Alergol. 40:447–461. 2023. View Article : Google Scholar : PubMed/NCBI

275 

Paoli A, Cenci L, Pompei P, Sahin N, Bianco A, Neri M, Caprio M and Moro T: Effects of two months of very low carbohydrate ketogenic diet on body composition, muscle strength, muscle area, and blood parameters in competitive natural body builders. Nutrients. 13:3742021. View Article : Google Scholar : PubMed/NCBI

276 

Ying W, Riopel M, Bandyopadhyay G, Dong Y, Birmingham A, Seo JB, Ofrecio JM, Wollam J, Hernandez-Carretero A, Fu W, et al: Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in vitro insulin sensitivity. Cell. 171:372–384.e12. 2017. View Article : Google Scholar

277 

Gao X, Chen Y, Wang J, Xu J, Wan H, Li X and Shi Y: Mitochondria-Rich extracellular vesicles from bone marrow stem cells mitigate muscle degeneration in rotator cuff tears in a rat model through macrophage M2 phenotype conversion. Arthroscopy. 41:3487–3499. 2025. View Article : Google Scholar : PubMed/NCBI

278 

Trentini M, D'Amora U, Ronca A, Lovatti L, Calvo-Guirado JL, Licastro D, Monego SD, Delogu LG, Wieckowski MR, Barak S, et al: Bone regeneration revolution: Pulsed electromagnetic field modulates macrophage-derived exosomes to attenuate osteoclastogenesis. Int J Nanomedicine. 19:8695–8707. 2024. View Article : Google Scholar : PubMed/NCBI

279 

Hu XM, Wang CC, Xiao Y, Liu Y, Huang HR, Jiang P, Wang YK, Lin YJ, Li LC and Qi ZQ: Non-clinical safety evaluation of exosomes derived from human umbilical cord mesenchymal stem cells in cynomolgus monkeys. Int J Nanomedicine. 19:4923–4939. 2024. View Article : Google Scholar : PubMed/NCBI

280 

Li JJ, Wang B, Kodali MC, Chen C, Kim E, Patters BJ, Lan L, Kumar S, Wang X, Yue J and Liao FF: In vivo evidence for the contribution of peripheral circulating inflammatory exosomes to neuroinflammation. J Neuroinflammation. 15:82018. View Article : Google Scholar : PubMed/NCBI

281 

Wang Y, Kong Y, Du J, Qi L, Liu M, Xie S, Hao J, Li M, Cao S, Cui H, et al: Injection of human umbilical cord mesenchymal stem cells exosomes for the treatment of knee osteoarthritis: From preclinical to clinical research. J Transl Med. 23:6412025. View Article : Google Scholar : PubMed/NCBI

282 

Muraca M and Cappariello A: The role of extracellular vesicles (EVs) in the epigenetic regulation of bone metabolism and osteoporosis. Int J Mol Sci. 21:86822020. View Article : Google Scholar : PubMed/NCBI

283 

Rohm TV, Castellani Gomes Dos Reis F, Isaac R, Murphy C, Rocha KCE, Bandyopadhyay G, Gao H, Libster AM, Zapata RC, Lee YS, et al: Adipose tissue macrophages secrete small extracellular vesicles that mediate rosiglitazone-induced insulin sensitization. Nat Metab. 6:880–898. 2024. View Article : Google Scholar : PubMed/NCBI

284 

Ying W, Gao H, Dos Reis FCG, Bandyopadhyay G, Ofrecio JM, Luo Z, Ji Y, Jin Z, Ly C and Olefsky JM: MiR-690, an exosomal-derived miRNA from M2-polarized macrophages, improves insulin sensitivity in obese mice. Cell Metab. 33:781–790.e5. 2021. View Article : Google Scholar : PubMed/NCBI

285 

Wang Y, Zhang X, Wang J, Zhang Y, Ye Q, Wang Y, Fei D and Wang Q: Inflammatory periodontal ligament stem cells drive m1 macrophage polarization via exosomal miR-143-3p-Mediated regulation of PI3K/AKT/NF-κB signaling. Stem Cells. 41:184–199. 2023. View Article : Google Scholar

286 

Chen Y, Dong J, Li J, Li J, Lu Y, Dong W, Zhang D and Dang X: Engineered macrophage-derived exosomes via click chemistry for the treatment of osteomyelitis. J Mater Chem B. 12:10593–10604. 2024. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Cui R, Zheng M, Hong J, Wang Z, Cun Y, Gao S, Zhu Y, Yang Z and Liu M: <p>Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)</p>. Int J Mol Med 57: 67, 2026.
APA
Cui, R., Zheng, M., Hong, J., Wang, Z., Cun, Y., Gao, S. ... Liu, M. (2026). <p>Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)</p>. International Journal of Molecular Medicine, 57, 67. https://doi.org/10.3892/ijmm.2026.5738
MLA
Cui, R., Zheng, M., Hong, J., Wang, Z., Cun, Y., Gao, S., Zhu, Y., Yang, Z., Liu, M."<p>Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)</p>". International Journal of Molecular Medicine 57.3 (2026): 67.
Chicago
Cui, R., Zheng, M., Hong, J., Wang, Z., Cun, Y., Gao, S., Zhu, Y., Yang, Z., Liu, M."<p>Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)</p>". International Journal of Molecular Medicine 57, no. 3 (2026): 67. https://doi.org/10.3892/ijmm.2026.5738
Copy and paste a formatted citation
x
Spandidos Publications style
Cui R, Zheng M, Hong J, Wang Z, Cun Y, Gao S, Zhu Y, Yang Z and Liu M: <p>Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)</p>. Int J Mol Med 57: 67, 2026.
APA
Cui, R., Zheng, M., Hong, J., Wang, Z., Cun, Y., Gao, S. ... Liu, M. (2026). <p>Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)</p>. International Journal of Molecular Medicine, 57, 67. https://doi.org/10.3892/ijmm.2026.5738
MLA
Cui, R., Zheng, M., Hong, J., Wang, Z., Cun, Y., Gao, S., Zhu, Y., Yang, Z., Liu, M."<p>Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)</p>". International Journal of Molecular Medicine 57.3 (2026): 67.
Chicago
Cui, R., Zheng, M., Hong, J., Wang, Z., Cun, Y., Gao, S., Zhu, Y., Yang, Z., Liu, M."<p>Research progress on the effects of macrophage‑derived exosomes on muscle factors IGF‑1 and FGF‑2 mediating musculoskeletal crosstalk molecular signaling pathway on bone metabolism (Review)</p>". International Journal of Molecular Medicine 57, no. 3 (2026): 67. https://doi.org/10.3892/ijmm.2026.5738
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team