Abstract

Psychological stress has been associated with the onset of several diseases, including osteoporosis. However, the underlying pathogenic mechanism remains unknown, and effective therapeutic strategies are still unavailable. Growing evidence suggests that the sympathetic nervous system regulates bone homeostasis and vascular function under psychological stress, as well as the coupling of osteogenesis and angiogenesis in bone development, remodeling, and regeneration. Furthermore, extracellular vesicles (EVs), particularly mesenchymal stem cell extracellular vesicles (MSC–EVs), have emerged as prospecting therapies for stimulating angiogenesis and bone regeneration. We summarize the role of sympathetic regulation in bone homeostasis and vascular function in response to psychological stress and emphasize the relationship between vessels and bone. Finally, we suggest using MSC–EVs as a promising therapeutic method for treating osteoporosis in psychological stress.

Introduction

Psychological stress is a state of tension characterized by physiological, emotional, and behavioral changes due to adversity, perceived stress, or threat [1–3]. People are experiencing significantly increased psychological pressure and stress caused by negative life events, which seriously impact physical health as the pace of life and work continues to accelerate [4]. Under normal circumstances, the stress response system has robust basic activity and response capabilities, critical for happiness, task achievement, and appropriate social interaction. However, the excessive or insufficient activity of this system impairs development and growth, resulting in various behavioral changes and pathological conditions [5]. Increasing evidence suggests that chronic psychological stress affects brain function and disrupts the balance of the stress system, resulting in inflammatory diseases, cardiovascular diseases, obesity, diabetes, atherosclerosis, cancer, neurodegenerative diseases, and osteoporosis [6–12].

Osteoporosis is a systemic skeletal disease that causes reduced bone mass and deterioration of bone tissue microstructure, increasing the risk of fractures [13]. Osteoporosis affected over 12.6% of non-institutionalized Americans aged 50 and over in 2017–2018 [14]. In 2017, 20 million people aged 50 or older in Europe had osteoporosis, with 2.7 million suffering osteoporotic fractures, resulting in a total treatment cost of 37.5 million euros and a considerable economic burden on society [15]. An increasing body of evidence indicates a substantial correlation between chronic psychological stress and bone loss [16–18], with studies revealing a positive relationship between the risk of osteoporotic fractures and psychological stress [19]. Furthermore, individuals diagnosed with post-traumatic stress disorder (PTSD) are more likely to develop osteoporosis [20], and long-term chronic psychological stress leads to depression, which is related to reduced bone mineral density (BMD) compared to non-depressed individuals [21].

Although earlier research has suggested a tight relationship between osteoporosis and psychological stress, the underlying mechanism and effective treatment techniques and approaches for osteoporosis under chronic psychological stress remain unknown and unestablished. We summarize the correlation between psychological stress and osteoporosis and highlight the function of the sympathetic nervous system (SNS) in regulating bone homeostasis. We also discuss the role of vasculature in bone development, remodeling, and regeneration, along with the effects of SNS on vascular regulation in bone. Furthermore, we focus on emerging therapeutics of extracellular vesicles (EVs), particularly mesenchymal stem cell-derived EV (MSC–EVs). We detail the impacts and mechanisms underpinning the use of MSC–EVs in vascular regulation and bone regeneration and propose the MSC–EV engineering strategy to enhance their efficiency and achieve more precise targeting.

Psychological stress and osteoporosis

Response mechanisms and sympathetic nervous system effects under psychological stress

The neuroendocrine response to stress causes numerous metabolic and functional changes in multiple organs and cells to maintain individual homeostasis in the face of possible dangers. The hypothalamus-pituitary-adrenal (HPA) axis and the sympathetic-adrenal medullary (SAM) axis are the two most important neuroendocrine response axes [22]. The HPA axis is activated when the hypothalamus secretes corticotropin-releasing hormone (CRH), stimulating adrenocorticotropic hormone (ACTH) release from the pituitary. As a result, glucocorticoid (GC) production and secretion from the adrenal cortex are regulated. GCs play crucial roles in reacting to harmful stimuli, maintaining blood pressure and blood sugar levels, mobilizing lipids, fighting cell damage, and regulating inflammatory reactions. Sympathetic activation increases, and norepinephrine (NE) is released from sympathetic nerve terminals when SAM axis is activated (Fig. 1). This activates the adrenal medulla to rapidly generate large amounts of catecholamines, mobilizing and regulating respiration, metabolism, and blood circulation, resulting in a state of alertness in the body and ensuring that critical organs can meet stress demands [23–26].

Psychological stress-induced sympathoexcitation leads to bone loss.Activation of sympathetic nerves triggers the release of NE and NPY, which bind to osteoblast β-AR and NPY receptors, initiating the RANK-RANKL pathway and driving osteoclast differentiation, ultimately resulting in bone loss. NE, norepinephrine; β-AR, beta-adrenergic receptor; NPY, neuropeptide Y.
Figure 1.

Psychological stress-induced sympathoexcitation leads to bone loss.Activation of sympathetic nerves triggers the release of NE and NPY, which bind to osteoblast β-AR and NPY receptors, initiating the RANK-RANKL pathway and driving osteoclast differentiation, ultimately resulting in bone loss. NE, norepinephrine; β-AR, beta-adrenergic receptor; NPY, neuropeptide Y.

The SNS is widely distributed, with postganglionic fibers from neurons in the paravertebral ganglia innervating the pupils, heart, respiratory system, blood vessels, facial and jaw muscles, trunk, and exocrine glands of the limbs. At the same time, postganglionic fibers from neurons in the prevertebral ganglia innervate the organs in the abdomen, pelvis, and perineum [27]. NE is the main SNS neurotransmitter, transmitting signals through adrenergic receptors (ARs), which are classified into α and β types and are both G protein-coupled receptors [28]. Dysfunction of the SNS is closely related to the onset and progression of many diseases, such as peripheral neuropathy, congestive heart failure, hypertension, diabetes, and immune dysfunction [29–33].

Regulation of bone homeostasis under psychological stress

Bones are critical organs that maintain posture and mobility, protect vital organs, and serve as crucial sites for hematopoiesis and mineral storage, in addition to playing an essential role in endocrine regulation and homeostasis [34, 35]. In the early 21st century, cross-sectional investigations on bone mass, body weight, and gonadal function have revealed new insights into how the nervous system regulates bone remodeling. Leptin was discovered in 2002 to alter SNS tone by acting on the hypothalamus, resulting in the release of NE into the local microenvironment and the activation of β-adrenergic receptors (β-ARs) produced by osteoblasts to modulate bone mass [36]. Additionally, neuropeptide U also regulates bone mass by signal processing through the SNS [37]. As research on the local microenvironment of bone progresses, the crucial regulatory role of SNS in bone metabolism has gradually emerged. The SNS is widely distributed in the periosteum and bone, densely distributed in the growth plate and metaphysis of long bones, and runs parallel to blood vessels. ARs and neuropeptide receptors have also been detected in osteoblasts and osteoclasts [38–40]. These findings imply that the SNS regulates the role of bone tissue.

Regulation of bone homeostasis is critical for adapting to changes in the physiology and external environment while maintaining bone morphology and function. This is mostly accomplished through bone remodeling, which involves the absorption of existing bone and the production of new bone, as conducted by osteoclasts and osteoblasts, respectively (Fig. 1). The balance between osteoblast and osteoclast activity maintains normal bone homeostasis [41]. The process of bone remodeling is mediated by the κ-β nuclear receptor (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) axis. RANKL released by cells in the osteoblast lineage binds to its receptor RANK on precursor cells of osteoclasts. Then, RANK-RANKL binding drives precursor differentiation of precursor cells into osteoclasts, which merge to produce mature multinucleated osteoclasts.

Furthermore, osteoblasts release OPG, which functions as a decoy receptor for RANKL and inhibits RANK-RANKL interaction, reducing osteoclastogenesis [42]. Psychological stress activates the SNS. NE produced by nerve fibers activates β-ARs on osteoblasts, causing them to create interleukin 6 (IL-6) and interleukin 11 (IL-11), inhibiting bone formation. It also induces osteoblast lineage cells to produce RANKL, promoting osteoclast differentiation [43–45]. Neuropeptide Y (NPY), frequently generated with NE, negatively regulates bone development and contributes to bone loss [46, 47].

In addition to directly regulating bone homeostasis through neurotransmitter release, the SNS also exerts indirect control on bone via paracrine mechanisms involving EVs and cytokines. Sui et al. demonstrated that the application of isoproterenol (ISO), a beta-adrenergic receptor agonist, induced osteoblasts to transport EVs containing miR-21 to osteoclasts [48]. Subsequently, miR-21 played a crucial role in modulating osteoclastogenesis in vivo by inhibiting programmed cell death 4 (Pdcd4), resulting in bone loss, not through a direct reduction in osteoblast activity [49]. Elefteriou et al. revealed that sympathetic activation induced by leptin triggers bone resorption by upregulating the expression of the osteoclast differentiation factor RANKL in osteoblast progenitor cells [50]. This process is mediated through phosphorylated activating transcription factor 4 (ATF4), which plays an essential role in osteoblast differentiation and function, rather than via direct regulation of osteoclasts. Considering the indirect regulatory role of the SNS in bone homeostasis, various therapeutic strategies have been proposed. For instance, targeted delivery approaches specifically designed to silence osteoblastic miR-21 or employing (D-Asp8)-lipid nanoparticle-mediated targeted inhibition of osteoclastic miR-21, as well as clinically relevant drugs, show promise in restoring the balance of bone remodeling and alleviating ISO- and CVS-induced osteoporosis [48, 49]. Additionally, considering the significance of ADRB2, an effective, bone-specific pharmacological blockade targeting ADRB2 signaling would be highly beneficial in mitigating bone loss induced by sympathetic activation.

Models of psychological stress and osteoporosis

Since the groundbreaking research by Holmes and Rahe in 1967, exploring the impact of life events on health, there has been a focused effort to establish precise animal models dedicated to studying the effects of psychological stress [51]. Currently, animal research models related to psychological stress are categorized into acute stress models, chronic stress models, and drug-induced models. Acute stress models, such as acute restraint stress and single prolonged stress, are designed to simulate situations in humans where experiences trigger intense negative emotions and a sense of trauma-inducing tension. These symptoms usually develop rapidly within minutes or hours–a quick reaction [52, 53]. Conversely, chronic stress models like social defeat, chronic variable stress, and trauma witness aim to simulate chronic changes in cognitive, emotional, physical, and behavioral aspects caused by a consistent sense of feeling pressured and overwhelmed over an extended period [52, 53].

Additionally, psychological stress models can also be established by administering drugs such as ISO and corticosterone (CORT) to mimic changes in hormones during psychological stress. The widespread use of this method is attributed to its singular treatment factor and predictable model establishment outcomes. Specific establishing methods of psychological stress related models have been summarized in Table 1. Moreover, the success of model establishment can be assessed through detections such as serum biomarker analysis, sucrose preference test, feeding experiments, social interaction trials, open field tests, forced swim tests, etc [48, 49, 79–83]. Among these psychological stress models, several have been confirmed to induce osteoporosis. For instance, in the study by Sui et al., models inducing psychological stress-related bone loss were established using chronic variable stress and ISO induction methods [48, 49]. Similarly, Zhang et al. detected reduced bone mass in the psychological stress model established through chronic social defeat stress methods [67].

Table 1.

Rodent models of psychological stress

ModelsMethodsDetections of sympathetic activationBone lossRef.
Time-dependent sensitization (TDS)Exposure to brief, intense stimuli followed by subsequent pharmacological and non-pharmacological stressors.Not enumeratedNot investigated[54]
Electric shock (ES)Inescapable electric foot shocks (shock duration and current depend on frequency), sometimes combined with situational reminders.Open-field test, noise test, elevated plus-maze testNot investigated[55–57]
Underwater trauma (UT)One-minute forced swim followed by 20–45 seconds of forced submersion in a water tank.Transfer testNot investigated[58, 59].
Restraint stress (RS)Immobilization on wooden boards or in a restraint container (e.g. syringe, wire mesh tube, 50 mL centrifuge tube) for 1–2 hours.Elevated plus-maze test, biomarker assaysYes[60–63]
Single prolonged stress (SPS)Two-hour restraint followed by a 20-minute forced swim and diethyl ether anesthesia until loss of consciousness.Biomarker assaysNot investigated[64]
Predator-based psychosocial stress (PPS)Exposure to a natural predator or predator-related stimuli, sometimes coupled with unstable housing conditions for 3–4 weeks.Elevated plus-maze test, noise test, radial-arm water maze, novel object recognition, biomarker assaysNot investigated[65, 66]
Social defeat (SD)Conspecific trained aggressor exposure for 6 hours daily for 5 or 10 days.Biomarker assays, open-field test, forced swim test, sucrose preference test, tail suspension test, partition test ethogram evaluation, urine marking testYes[67, 68]
Social isolation (SI)Social isolation (complete or near-complete lack of social interaction) during 3–4 weeks.Training test, contextual test, cued test, extinction testYes[69, 70]
Trauma witness (TW)Witnessing the social defeat event three times in one week.Open-field test, elevated plus-maze test, light-dark exploration, forced swim stress, sucrose preference test, memory function testNot investigated[71]
Early life stress (ELS)Separating animal pups from mothers for several hours daily during postnatal days 1–10, followed by re-exposure to stressors (e.g. underwater trauma) in adulthood.Biomarker assays, elevated plus-maze test, acoustic startle response test, ultrasonic vocalizations, open-field testYes[72–74]
Tail suspension (TS)Suspension by the tail for 2–4 weeks.Not enumeratedYes[75, 76]
Chronic variable stress (CVS)Exposure to variable stressors for 5–9 weeks, e.g. overnight illumination, changing cage mates, food deprivation and so on.Biomarker assays, sucrose preference testYes[48, 49]
Chronic subordinate colony housing paradigm (CSC)Housing in prolonged subordination to a dominant resident mouse for 19 consecutive days.Biomarker assays, elevated plus-maze testNot investigated[77]
Drug inductionDaily intraperitoneal injections of ISO solution (10 mg/kg) for 4 weeks.Biomarker assaysYes[48, 49]
Daily subcutaneous injections of CORT solution (20 mg/kg) for 6 weeks.Tail suspension test, forced swimming test, open field testNot investigated[78]
ModelsMethodsDetections of sympathetic activationBone lossRef.
Time-dependent sensitization (TDS)Exposure to brief, intense stimuli followed by subsequent pharmacological and non-pharmacological stressors.Not enumeratedNot investigated[54]
Electric shock (ES)Inescapable electric foot shocks (shock duration and current depend on frequency), sometimes combined with situational reminders.Open-field test, noise test, elevated plus-maze testNot investigated[55–57]
Underwater trauma (UT)One-minute forced swim followed by 20–45 seconds of forced submersion in a water tank.Transfer testNot investigated[58, 59].
Restraint stress (RS)Immobilization on wooden boards or in a restraint container (e.g. syringe, wire mesh tube, 50 mL centrifuge tube) for 1–2 hours.Elevated plus-maze test, biomarker assaysYes[60–63]
Single prolonged stress (SPS)Two-hour restraint followed by a 20-minute forced swim and diethyl ether anesthesia until loss of consciousness.Biomarker assaysNot investigated[64]
Predator-based psychosocial stress (PPS)Exposure to a natural predator or predator-related stimuli, sometimes coupled with unstable housing conditions for 3–4 weeks.Elevated plus-maze test, noise test, radial-arm water maze, novel object recognition, biomarker assaysNot investigated[65, 66]
Social defeat (SD)Conspecific trained aggressor exposure for 6 hours daily for 5 or 10 days.Biomarker assays, open-field test, forced swim test, sucrose preference test, tail suspension test, partition test ethogram evaluation, urine marking testYes[67, 68]
Social isolation (SI)Social isolation (complete or near-complete lack of social interaction) during 3–4 weeks.Training test, contextual test, cued test, extinction testYes[69, 70]
Trauma witness (TW)Witnessing the social defeat event three times in one week.Open-field test, elevated plus-maze test, light-dark exploration, forced swim stress, sucrose preference test, memory function testNot investigated[71]
Early life stress (ELS)Separating animal pups from mothers for several hours daily during postnatal days 1–10, followed by re-exposure to stressors (e.g. underwater trauma) in adulthood.Biomarker assays, elevated plus-maze test, acoustic startle response test, ultrasonic vocalizations, open-field testYes[72–74]
Tail suspension (TS)Suspension by the tail for 2–4 weeks.Not enumeratedYes[75, 76]
Chronic variable stress (CVS)Exposure to variable stressors for 5–9 weeks, e.g. overnight illumination, changing cage mates, food deprivation and so on.Biomarker assays, sucrose preference testYes[48, 49]
Chronic subordinate colony housing paradigm (CSC)Housing in prolonged subordination to a dominant resident mouse for 19 consecutive days.Biomarker assays, elevated plus-maze testNot investigated[77]
Drug inductionDaily intraperitoneal injections of ISO solution (10 mg/kg) for 4 weeks.Biomarker assaysYes[48, 49]
Daily subcutaneous injections of CORT solution (20 mg/kg) for 6 weeks.Tail suspension test, forced swimming test, open field testNot investigated[78]
Table 1.

Rodent models of psychological stress

ModelsMethodsDetections of sympathetic activationBone lossRef.
Time-dependent sensitization (TDS)Exposure to brief, intense stimuli followed by subsequent pharmacological and non-pharmacological stressors.Not enumeratedNot investigated[54]
Electric shock (ES)Inescapable electric foot shocks (shock duration and current depend on frequency), sometimes combined with situational reminders.Open-field test, noise test, elevated plus-maze testNot investigated[55–57]
Underwater trauma (UT)One-minute forced swim followed by 20–45 seconds of forced submersion in a water tank.Transfer testNot investigated[58, 59].
Restraint stress (RS)Immobilization on wooden boards or in a restraint container (e.g. syringe, wire mesh tube, 50 mL centrifuge tube) for 1–2 hours.Elevated plus-maze test, biomarker assaysYes[60–63]
Single prolonged stress (SPS)Two-hour restraint followed by a 20-minute forced swim and diethyl ether anesthesia until loss of consciousness.Biomarker assaysNot investigated[64]
Predator-based psychosocial stress (PPS)Exposure to a natural predator or predator-related stimuli, sometimes coupled with unstable housing conditions for 3–4 weeks.Elevated plus-maze test, noise test, radial-arm water maze, novel object recognition, biomarker assaysNot investigated[65, 66]
Social defeat (SD)Conspecific trained aggressor exposure for 6 hours daily for 5 or 10 days.Biomarker assays, open-field test, forced swim test, sucrose preference test, tail suspension test, partition test ethogram evaluation, urine marking testYes[67, 68]
Social isolation (SI)Social isolation (complete or near-complete lack of social interaction) during 3–4 weeks.Training test, contextual test, cued test, extinction testYes[69, 70]
Trauma witness (TW)Witnessing the social defeat event three times in one week.Open-field test, elevated plus-maze test, light-dark exploration, forced swim stress, sucrose preference test, memory function testNot investigated[71]
Early life stress (ELS)Separating animal pups from mothers for several hours daily during postnatal days 1–10, followed by re-exposure to stressors (e.g. underwater trauma) in adulthood.Biomarker assays, elevated plus-maze test, acoustic startle response test, ultrasonic vocalizations, open-field testYes[72–74]
Tail suspension (TS)Suspension by the tail for 2–4 weeks.Not enumeratedYes[75, 76]
Chronic variable stress (CVS)Exposure to variable stressors for 5–9 weeks, e.g. overnight illumination, changing cage mates, food deprivation and so on.Biomarker assays, sucrose preference testYes[48, 49]
Chronic subordinate colony housing paradigm (CSC)Housing in prolonged subordination to a dominant resident mouse for 19 consecutive days.Biomarker assays, elevated plus-maze testNot investigated[77]
Drug inductionDaily intraperitoneal injections of ISO solution (10 mg/kg) for 4 weeks.Biomarker assaysYes[48, 49]
Daily subcutaneous injections of CORT solution (20 mg/kg) for 6 weeks.Tail suspension test, forced swimming test, open field testNot investigated[78]
ModelsMethodsDetections of sympathetic activationBone lossRef.
Time-dependent sensitization (TDS)Exposure to brief, intense stimuli followed by subsequent pharmacological and non-pharmacological stressors.Not enumeratedNot investigated[54]
Electric shock (ES)Inescapable electric foot shocks (shock duration and current depend on frequency), sometimes combined with situational reminders.Open-field test, noise test, elevated plus-maze testNot investigated[55–57]
Underwater trauma (UT)One-minute forced swim followed by 20–45 seconds of forced submersion in a water tank.Transfer testNot investigated[58, 59].
Restraint stress (RS)Immobilization on wooden boards or in a restraint container (e.g. syringe, wire mesh tube, 50 mL centrifuge tube) for 1–2 hours.Elevated plus-maze test, biomarker assaysYes[60–63]
Single prolonged stress (SPS)Two-hour restraint followed by a 20-minute forced swim and diethyl ether anesthesia until loss of consciousness.Biomarker assaysNot investigated[64]
Predator-based psychosocial stress (PPS)Exposure to a natural predator or predator-related stimuli, sometimes coupled with unstable housing conditions for 3–4 weeks.Elevated plus-maze test, noise test, radial-arm water maze, novel object recognition, biomarker assaysNot investigated[65, 66]
Social defeat (SD)Conspecific trained aggressor exposure for 6 hours daily for 5 or 10 days.Biomarker assays, open-field test, forced swim test, sucrose preference test, tail suspension test, partition test ethogram evaluation, urine marking testYes[67, 68]
Social isolation (SI)Social isolation (complete or near-complete lack of social interaction) during 3–4 weeks.Training test, contextual test, cued test, extinction testYes[69, 70]
Trauma witness (TW)Witnessing the social defeat event three times in one week.Open-field test, elevated plus-maze test, light-dark exploration, forced swim stress, sucrose preference test, memory function testNot investigated[71]
Early life stress (ELS)Separating animal pups from mothers for several hours daily during postnatal days 1–10, followed by re-exposure to stressors (e.g. underwater trauma) in adulthood.Biomarker assays, elevated plus-maze test, acoustic startle response test, ultrasonic vocalizations, open-field testYes[72–74]
Tail suspension (TS)Suspension by the tail for 2–4 weeks.Not enumeratedYes[75, 76]
Chronic variable stress (CVS)Exposure to variable stressors for 5–9 weeks, e.g. overnight illumination, changing cage mates, food deprivation and so on.Biomarker assays, sucrose preference testYes[48, 49]
Chronic subordinate colony housing paradigm (CSC)Housing in prolonged subordination to a dominant resident mouse for 19 consecutive days.Biomarker assays, elevated plus-maze testNot investigated[77]
Drug inductionDaily intraperitoneal injections of ISO solution (10 mg/kg) for 4 weeks.Biomarker assaysYes[48, 49]
Daily subcutaneous injections of CORT solution (20 mg/kg) for 6 weeks.Tail suspension test, forced swimming test, open field testNot investigated[78]

Currently, osteoporosis is categorized into primary and secondary types. Primary osteoporosis involves gonadectomy and aging, while secondary osteoporosis includes conditions such as diabetes, drug induction, disuse, psychological stress, and radiation exposure [84]. Table 2 outlines specific methodologies used to create models related to osteoporosis. Notably, some of these methods have been observed to trigger sympathetic activation (Table 2). For instance, Sui et al. uncovered that various osteoporotic mouse models such as ovariectomized mice and aging mice exhibited a significant increase in serum levels of the sympathetic activation marker–NE, but the mechanism remains unclear [48]. Moreover, the intricate distribution of nerves, blood vessels, and other components in bone, along with their interactions, remains a crucial area for investigation. In appropriate animal models, further elucidating whether and how the SNS regulates bone homeostasis through influencing blood vessels and other structures is of paramount importance in better understanding the relationship between psychological stress and osteoporosis.

Table 2.

Rodent models of osteoporosis

ModelsMethodsSympathetic activationRef.
GonadectomyOvariectomy (OVX) or orchidectomy (ORX).Yes[48]
AgingNatural aging or using senescence accelerated-prone mice.
Drug inductionDaily intraperitoneal injection of dexamethasone (20 mg/kg) for 4 weeks.
DisuseTail suspension or hindlimb immobilization for 2 weeks.
Psychological stressExposure to various stressors or daily ISO induction (10 mg/kg) for 4 weeks.
Type 1 DiabetesDaily intraperitoneal injection of streptozotocin (50 mg/kg) for 5 days.
Type 2 DiabetesInduction of insulin resistance via high-fat diet or other means.Yes[85]
RadiationFocal radiation as a single dose of 20 Gy.Not investigated[86]
ModelsMethodsSympathetic activationRef.
GonadectomyOvariectomy (OVX) or orchidectomy (ORX).Yes[48]
AgingNatural aging or using senescence accelerated-prone mice.
Drug inductionDaily intraperitoneal injection of dexamethasone (20 mg/kg) for 4 weeks.
DisuseTail suspension or hindlimb immobilization for 2 weeks.
Psychological stressExposure to various stressors or daily ISO induction (10 mg/kg) for 4 weeks.
Type 1 DiabetesDaily intraperitoneal injection of streptozotocin (50 mg/kg) for 5 days.
Type 2 DiabetesInduction of insulin resistance via high-fat diet or other means.Yes[85]
RadiationFocal radiation as a single dose of 20 Gy.Not investigated[86]
Table 2.

Rodent models of osteoporosis

ModelsMethodsSympathetic activationRef.
GonadectomyOvariectomy (OVX) or orchidectomy (ORX).Yes[48]
AgingNatural aging or using senescence accelerated-prone mice.
Drug inductionDaily intraperitoneal injection of dexamethasone (20 mg/kg) for 4 weeks.
DisuseTail suspension or hindlimb immobilization for 2 weeks.
Psychological stressExposure to various stressors or daily ISO induction (10 mg/kg) for 4 weeks.
Type 1 DiabetesDaily intraperitoneal injection of streptozotocin (50 mg/kg) for 5 days.
Type 2 DiabetesInduction of insulin resistance via high-fat diet or other means.Yes[85]
RadiationFocal radiation as a single dose of 20 Gy.Not investigated[86]
ModelsMethodsSympathetic activationRef.
GonadectomyOvariectomy (OVX) or orchidectomy (ORX).Yes[48]
AgingNatural aging or using senescence accelerated-prone mice.
Drug inductionDaily intraperitoneal injection of dexamethasone (20 mg/kg) for 4 weeks.
DisuseTail suspension or hindlimb immobilization for 2 weeks.
Psychological stressExposure to various stressors or daily ISO induction (10 mg/kg) for 4 weeks.
Type 1 DiabetesDaily intraperitoneal injection of streptozotocin (50 mg/kg) for 5 days.
Type 2 DiabetesInduction of insulin resistance via high-fat diet or other means.Yes[85]
RadiationFocal radiation as a single dose of 20 Gy.Not investigated[86]

Vascular regulation and bone homeostasis

Vasculature coupled with bone formation and regeneration

Blood vessels are widely distributed in tissues and organs and perform the physiological function of blood transport and substance exchange by joining arteries, capillaries, and veins in sequence as a continuous and relatively closed tube system. Blood vessels also have a role in the physiological and pathological processes of bone, such as skeletal development, regeneration, hematopoiesis, and homeostasis regulation [87]. The connection between vascularization and osteogenesis is illustrated by endochondral ossification. Hypertrophic chondrocytes (HCs) begin hypoxia-inducible factor (HIF) signaling, enhance the expression of vascular endothelial growth factor (VEGF) target genes, and promote blood vessel formation [88] in the primary ossification center (POC). Matrix metalloproteinases (MMPs) are secreted by osteoclasts and endothelial cells (ECs), which degrade the extracellular matrix (ECM) and increase VEGF signaling. VEGF-mediated signaling promotes migration and proliferation of these cell types [89, 90] when ECs and osteoblasts express VEGFR2.

Researchers are increasingly interested in the relationship between angiogenesis and osteogenesis. Kusumbe et al. discovered in 2014 that different vascular types exhibit heterogeneity of ECs in bone. ECs in the metaphysis and endosteum had high expressions of CD31 and endomucin (EMCN) (CD31hiEMCNhi), identifying them as type H vessels. At the same time, ECs in the bone shaft had low CD31 and EMCN (CD31loEMCNlo) expressions, identifying them as type L vessels [91]. Type H vessels are in tight contact with osteoprogenitor cells to provide necessary nutrients for bone formation, satisfy metabolic demands of bone development, and maintain bone homeostasis [92–94]. The expression levels of platelet-derived growth factor (PDGF), transforming growth factor (TGF), and other cytokines related to osteoprogenitor cell survival and proliferation were significantly higher in type H endothelium than in type L endothelium. Furthermore, in the metaphysis and endosteum, MSCs positive for platelet-derived growth factor receptor β (PDGFRβ), osteoblast-lineage cells positive for runt-related transcription factor 2 (RUNX2), and osteoblast-specific transcription factor (OSX) aggregate with type H endothelium. As a result, they exhibit a substantial spatial correlation, demonstrating the maintenance effect of type H endothelium on osteoblast-lineage cells.

Vascular regulation by sympathetic nervous system

The regulation of angiotasis requires the intricate integration of several signals that promote vasodilation and vasoconstriction to maintain blood pressure and volume, ensuring appropriate oxygen supply to metabolically active tissues. There is a state of tensional balance between the vasoactive substances released by the endothelium and the neurotransmitters produced by sympathetic nerve terminals [95]. The endothelium produces and releases numerous vasoactive substances in normal conditions, including endothelin 1 (ET-1) and angiotensin II (Ang II) for vasoconstriction and nitric oxide (NO), prostacyclin (PGI2), and endothelium-derived hyperpolarizing factor (EDHF) for vasodilation [96]. Under sympathoexcitation, the released NE causes systemic vasoconstriction and increases blood pressure by causing smooth muscle contraction via increasing the intercellular concentration of Ca2+ (Fig. 2). The endothelium inhibits sympathetic activity by promoting the metabolism and breakdown of NE to relieve vasoconstriction, providing a physical barrier to prevent NE diffusion into the bloodstream [97]. Moreover, in the coronary and large arteries, the activation of α2-ARs and β-ARs expressed on ECs leads to NO release and further vasodilation [98].

Sympathetic activation governs various aspects of vascular function.Sympathetic activation influences vascular vasoconstriction via Ca2+ concentration modulation. Endothelial functions, including adhesion, permeability, and inflammation, are regulated by the activation of α-ARs during sympathetic activation. Additionally, angiogenesis of endothelial progenitor cells (EPCs) is governed by β2-AR stimulation, which promotes EPC proliferation, migration, and differentiation. α-AR, alpha-adrenergic receptor; β2-AR, beta 2-adrenergic receptor; EPC, endothelial progenitor cell.
Figure 2.

Sympathetic activation governs various aspects of vascular function.Sympathetic activation influences vascular vasoconstriction via Ca2+ concentration modulation. Endothelial functions, including adhesion, permeability, and inflammation, are regulated by the activation of α-ARs during sympathetic activation. Additionally, angiogenesis of endothelial progenitor cells (EPCs) is governed by β2-AR stimulation, which promotes EPC proliferation, migration, and differentiation. α-AR, alpha-adrenergic receptor; β2-AR, beta 2-adrenergic receptor; EPC, endothelial progenitor cell.

The SNS has been found to not only affect vasoconstriction and vasodilation, but also influence endothelial function and angiogenesis through Ars, with the continuous deepening of research on neurovascular interactions (Fig. 2). Angiogenesis refers to the biological process of producing new blood vessels on existing vasculature, often by sprouting and intussusception, and it is critical in physiological and pathological processes [99, 100]. Intussusceptive angiogenesis is the process by which a single blood vessel divides to form two lumens, also known as splitting angiogenesis [101, 102]. Sprouting angiogenesis involves forming new blood vessels from existing ones by endothelial sprouting [103, 104]. Studies have shown that the activation of α-ARs leads to endothelial dysfunction characterized by increased white blood cell adhesion to ECs, vascular inflammation, and increased vascular permeability [105–107]. Furthermore, stimulating β2-ARs on endothelial progenitor cells (EPCs) leads to its proliferation, migration, differentiation, and angiogenesis [108].

Specialized neurovascular coupling regulated by sympathetic nervous system

Bone blood vessels are densely distributed and continually transport oxygen, nutrients, and waste. At the same time, they also play a critical role in physiological and pathological processes. The discovery of type H vessels confirms the heterogeneity of ECs and provides evidence supporting the theory of angiogenesis–osteogenesis coupling. Research on promoting bone formation by type H vessels is increasing, highlighting the importance of specialized vasculature in bone homeostasis and regeneration.

Notably, close spatial connections between sympathetic nerves and type H vessels in metaphysis have been proven, in which ADRBs, particularly ADRB2, negatively regulate type H vasculature and bone homeostasis (Fig. 3).

Neurovascular coupling and their regulation in bone homeostasis and regeneration under sympathetic activation.Close spatial connections and functional coupling exist between sympathetic nerves and type H vessels in the metaphysis. Sympathetic activation stimulates MSCs to transcriptionally regulate type H vessel angiogenesis via paracrine effects, subsequently impacting bone homeostasis and regeneration through the coupling of angiogenesis and osteogenesis.
Figure 3.

Neurovascular coupling and their regulation in bone homeostasis and regeneration under sympathetic activation.Close spatial connections and functional coupling exist between sympathetic nerves and type H vessels in the metaphysis. Sympathetic activation stimulates MSCs to transcriptionally regulate type H vessel angiogenesis via paracrine effects, subsequently impacting bone homeostasis and regeneration through the coupling of angiogenesis and osteogenesis.

Sympathoexcitation induces a reduction in type H vessels and bone mass in vivo, primarily through indirect sympathetic modulation of angiogenesis mediated by the paracrine effects of mesenchymal stem cells (MSCs). This regulatory mechanism involves alterations in the transcriptional activity of several angiogenic genes within ECs.

Xu et al. elucidated a distinct type of neurovascular coupling, proposing a novel approach centered on neural signaling and targeted vasculature for osteoanabolic therapies. Although the mechanisms by which pericytes respond to sympathetic activation and influence the formation of type H vessels through paracrine mechanisms remain unclear, this indirect regulatory mechanism may provide a new perspective for understanding neurovascular coupling in bone. EVs, as a crucial mode of intercellular signaling, have the capability to indirectly respond to sympathetic activation, thereby regulating bone homeostasis. Hence, investigating whether and how they participate in the sympathetic regulation of vasculature holds significant importance in further elucidating the mechanisms behind sympathoexcitation in regulating specialized type H endothelium and its potential effects on bone homeostasis and regeneration under psychological stress.

Extracellular vesicles and vascular regulation

Functions and applications of extracellular vesicles

EVs are small membrane-bound vesicles secreted actively by cells [109]. Initially, they were considered a form of waste excreted by cells. However, Harding and Pan discovered in 1983 that reticulocytes secreted small vesicles during maturation, transferring transferrin to the extracellular space [110, 111]. Later, Raposo et al. found that EVs secreted by B lymphocytes activated T lymphocytes and participated in immune regulation [112]. Since then, the functions of EVs have gained more attention. EVs are categorized according to their size and are distinguished as exosomes (40–160 nm), microvesicles (100–1000 nm), and apoptotic bodies (1000–5000 nm) [113, 114]. EVs remove excess or unnecessary components from cells to maintain cellular homeostasis and play a vital role in regulating intercellular communication by carrying proteins, nucleic acids, small molecules, and structural signaling molecules [115–117]. The key and complex processes that mediate intercellular communication include EVs recognition, binding, cellular uptake, and intracellular transport. EVs possess integrins, ECM proteins, lectins, proteoglycans, and glycolipids, enabling them to recognize and bind to specific receptors on target cells [118]. Subsequently, EVs mainly deliver their contents to the interior of cells by binding to receptors on target cells, fusing with the target cell plasma membranes, or being internalized, exerting biological effects.

The potential of EVs for diagnosing and treating several diseases, including neurodegenerative diseases and cardiovascular diseases, has recently been revealed [119–122]. EVs are abundant in blood and other bodily fluids, and their amounts and cargo of nucleic acids, proteins, lipids, and other substances reflect the physiological and pathological processes of the body, prompting them to be examined as biomarkers for various diseases [123]. Regarding treatment, EVs include functional substances, such as exogenous drugs, therapeutic RNA molecules, or proteins. They have been changed on their surface to create biological effects on receptor cells, allowing them to be used as a functional therapy for specific diseases [124–126].

Angiogenesis regulation by EVs

EVs control several physiological and pathological processes by delivering bioactive molecules. Numerous studies have shown that EVs play a critical role in angiogenesis regulation. For example, EVs produced from ECs contain β1 integrin, MMP-2, and MMP-9, which promote EC invasion and capillary-like structure formation [127]. EVs derived from endothelial colony-forming cells (ECFCs), which possess clear endothelial differentiation potential and proliferation activity, engage with α4 and β1 integrins to activate angiogenesis by conveying mRNA from the eNOS and PI3K/Akt signaling pathways [128, 129]. Recently, EVs have been considered potential mediators of remote ischemic adaptation and general protective signals. The ability of EVs to regulate angiogenesis offers new therapeutic opportunities for regenerative medicine and tissue engineering [130, 131]. Intramyocardial treatment with EVs can increase the perfusion of ischemic myocardial tissue by inducing arteriole and capillary growth in chronic ischemic heart disease [132]. EVs derived from ECFCs prevented capillary rarefaction and subsequent tissue damage in a rat model of renal ischemia-reperfusion injury [133]. EVs secreted by ECFCs enhance angiogenesis and accelerate wound healing by strengthening blood vessels at the wound site [134].

Furthermore, MSC-EVs has been confirmed to promote the formation of type H vessels in bone. Zhuang et al. induced the proangiogenic activity of MSCs through a hypoxic microenvironment. They confirmed that miR-210-3p, upregulated in small EVs derived from hypoxic MSCs, downregulated EFNA3 expression and enhanced the phosphorylation of the PI3K/AKT pathway, which facilitated the proliferation, migration, and angiogenesis abilities of ECs and ultimately enhanced bone regeneration and type H vessel formation in the critical size calvarial bone defect model [135]. Not only in bone, MSC-EVs have also been proven to promote the formation of type H vessels in the skin. Liu et al. demonstrated that EVs derived from engineered mesenchymal stem/stromal cell aggregates enriched angiogenic proteins. These proteins activated the Notch-related signaling pathway, promoting the formation of type H vessels in diabetic wounds, and ultimately facilitating their healing [136].

In summary, EVs are intercellular communication mediators used for diagnosing and treating various diseases. EVs produced from blood-related cells or other sources have been demonstrated to influence angiogenesis and have effective therapeutic benefits. However, it is still unknown whether sympathetic under psychological stress affects the role of EVs in angiogenesis regulation. Type H vessels are tightly coupled with bone homeostasis and regeneration, and it is unclear how they respond to the regulation of endogenous EVs and the effects of exogenous EVs on type H vessel maintenance in the context of sympathetic activation. Therefore, it is critical to investigate the role of EVs in specialized regeneration-related vascular structure regulation under sympathetic activation to develop therapy methods for bone loss caused by psychological stress.

MSC-EVs regulate angiogenesis and bone regeneration

MSC-EVs in angiogenesis regulation

MSCs are a type of adult non-hematopoietic stem cell discovered by Friedenstein et al. in bone marrow. They possess characteristics such as adhesion to plastic, self-renewal, clonogenicity, and multipotential differentiation abilities [137, 138]. MSCs are isolated from various body parts, including blood, bone marrow, liver, umbilical cord, periodontal ligament, lung, and adipose tissue [139] (Fig. 4). During tissue regeneration, it was initially believed that MSCs exerted therapeutic effects by migrating to the injury site, engrafting, and subsequently differentiating into the cells required for tissue regeneration. However, increasing studies have shown that the therapeutic benefits of MSCs are attributed to their differentiation and the active substances they secrete, especially the role of EVs, which has received extensive attention [140–142]. MSC-EVs transmit information to damaged cells or tissues, participate in tissue regeneration and functional repair, regulate extracellular matrix remodeling, limit local inflammation, and regulate immune responses [143–145]. Numerous studies have found that EVs released by MSCs have therapeutic potential in repairing diseases and functional damage in various tissues and organs, such as the lung, kidney, liver, nervous system, bone, cartilage, and heart [146–152].

Regulation of angiogenesis and bone regeneration by MSC-EVs.MSCs are derived from various tissues, including the umbilical cord, bone marrow, liver, tooth, and lung. MSC-EVs encompass exosomes, microvesicles, and apoptotic bodies, containing diverse components like proteins and nucleic acids. MSC-EVs are employed for promoting bone regeneration and regulating angiogenesis. MSC, mesenchymal stem cells; MSC-EVs, mesenchymal stem cell extracellular vesicles.
Figure 4.

Regulation of angiogenesis and bone regeneration by MSC-EVs.MSCs are derived from various tissues, including the umbilical cord, bone marrow, liver, tooth, and lung. MSC-EVs encompass exosomes, microvesicles, and apoptotic bodies, containing diverse components like proteins and nucleic acids. MSC-EVs are employed for promoting bone regeneration and regulating angiogenesis. MSC, mesenchymal stem cells; MSC-EVs, mesenchymal stem cell extracellular vesicles.

Several studies have demonstrated the significant role of MSC-EVs in promoting angiogenesis [153, 154] (Fig. 4). For instance, under hypoxic stimulation, bone marrow-derived MSC-EVs are absorbed by ECs, thereby promoting angiogenesis and enhancing cardiac function in rat myocardial infarction models [155]. After low oxygen treatment, umbilical cord-derived MSC-EVs improve the formation of capillary-like structures and promote blood flow recovery in rat models of hindlimb ischemia [156, 157]. Moreover, umbilical cord-derived MSC-EVs overexpressing Akt stimulate EC migration, proliferation, and tube formation in vitro, increasing blood vessel formation in vivo [158]. Similarly, stem cells from human exfoliated deciduous teeth (SHED)-derived MSC-EVs mediate endothelial cell differentiation and enhance angiogenesis by transferring miR-26a via the TGF-β/SMAD2/3 signaling pathway [159].

MSC-EVs in bone regeneration

In recent years, multiple studies have confirmed that MSC-EVs directly promote bone cell proliferation, differentiation, and angiogenesis while inhibiting inflammatory responses in various pathological conditions [160–163] (Fig. 4). Treatment in the osteonecrosis model with MSC–EV increased trabecular bone cell numbers, decreased apoptotic cell numbers, and reduced adipose tissue [164]. MSC-EVs in the fracture model promoted bone callus formation and enhanced bone healing [165]. Profound remodeling of newly formed bone in the osteoporosis model was observed with MSC-EVs treatment. In contrast, the control group without MSC-EVs treatment exhibited bone loss or minimal mineralization and bone formation [166]. Furthermore, MSC-EVs promote angiogenesis in osteoporotic rats, which is beneficial for bone formation [163]. More experiments have also revealed the potential mechanism of MSC-EVs in bone regeneration therapy. MSC-EVs may promote the osteogenic differentiation of bone marrow MSCs and induce osteogenesis by regulating the PI3K/Akt signaling pathway [167]. Additionally, MSC-EVs stimulate the proliferation and differentiation of osteoblasts by regulating the BMP/Smad/Wnt signaling pathway, thereby promoting osteogenesis [165, 166].

Although MSC-EVs have shown great potential in regenerative therapy, their application is limited by low natural production. In recent years, engineering techniques have been developed to modify MSC-EVs to avoid potential pathogenic risks and enhance their efficacy and production to meet the demand for MSC-EVs in diseases [167]. Engineering MSC-EVs involves surface modification and content loading. Surface modification is achieved through cosmetic cell modification and non-covalent or covalent membrane binding to make MSC-EVs target specific cells or reduce the probability of being cleared [168]. Loading functional components, including nucleic acids, proteins, and small molecules, into the lipid membrane bilayer structure on the surface of MSC-EVs and the hydrophilic core surrounded by the lipid membrane is known as content loading. Passive loading involves co-incubating functional therapeutic substances with EVs or donor cells, whereas active loading involves ultrasonic treatment, extrusion procedures, electroporation, multiple freeze-thaw, and other techniques [152, 169, 170]. While passive loading is straightforward and maintains the EV membranes intact, it has poor loading efficiency. On the other hand, active loading can provide a high loading efficiency for hydrophilic or macromolecular compounds but might result in issues, such as damage to membrane integrity or condensation [171–173].

Overall, MSC-EVs derived from MSCs with multipotent differentiation potential promote tissue regeneration, limit local inflammation, regulate immune responses, and effectively promote angiogenesis and bone regeneration. However, it is unclear how MSC-EVs respond to and exert their regenerative-promoting effects in the overall environment under sympathetic activation. The low natural production and lack of specificity of MSC-EVs limit their broad application in regenerative medicine. Therefore, engineering modifications of MSC-EVs to improve their targeting and therapeutic effects will better meet the demand for MSC-EVs in bone regeneration. Modified MSC-EVs with proangiogenic and pro-osteogenic effects will provide new options and directions for treating bone loss under psychological stress.

Summary and prospectives

Psychological stress is closely related to the development and progression of several diseases, including bone loss and osteoporosis. SNS, one of the neuroendocrine foundations of psychological stress, has been shown to significantly contribute to this process via overactivation or malfunction. However, the mechanism underlying sympathetic activation-induced bone loss remains unknown, and no viable therapy strategies are available. Recent research suggests sympathetic activation modulates bone and vascular cell homeostasis by acting on bone cells and ECs. Notably, the close distribution and mutual interaction of nerves and blood vessels in bone suggest that sympathetic activation might participate in bone regeneration by influencing vascular function. Identifying type H vessels, a specialized vasculature linked to bone regeneration highlights the significance of vascularization in modulating the response to sympathetic activation-induced bone loss. An emerging therapy based on EVs, namely MSC–EVs, is proposed to repair bone loss under psychological stress, with favorable proangiogenic and pro-osteogenic effects. We also propose engineering modifications to improve the targeting and therapeutic benefits of MSC-EVs to develop an effective novel technique for treating osteoporosis under psychological stress.

Acknowledgements

This work is supported by grants from the National Natural Science Foundation of China (Nos. 82100969, 81930025, 82371020 and 82301028), the Key research and Development Program of Shaanxi Province (No. 2023-YBSF-189), the Natural Science Basic Research Program of Shaanxi Province (No. 2022JQ-929), the Special Project of National Clinical Research Center for Oral Diseases (No. LCB202006), the Shaanxi Province Innovation Capability Support Program Scientific and Technological Innovation Team (No. 2023-CX-TD-69), the National Defense Biotechnology Outstanding Young Talents Fund (No. 01-SWKJYCJJ24), the “Rapid Response” Research projects (No. 2023KXKT017), the China Postdoctoral Science Foundation (No. BX20230485) and the Young Science and Technology Rising Star Project of Shaanxi Province (No. 2023KJXX-027). Figures 1 to 4 were modified from Servier Medical Art (smart.servier.com), licensed under a Creative Common Attribution 3.0 Generic License (creativecommons.org/licenses/by/3.0).

Author contributions

Hao-Kun Xu (Funding acquisition [Supporting], Writing—original draft [Equal], Writing—review & editing [Supporting]), Jie-Xi Liu (Writing—original draft [Equal], Writing—review & editing [Supporting]), Ze-Kai Zhou (Writing—original draft [Equal]), Chen-Xi Zheng (Writing—review & editing [Supporting]), Bing-Dong Sui (Writing—review & editing [Supporting]), Yuan Yuan (Writing—original draft [Supporting]), Liang Kong (Funding acquisition [Supporting], Supervision [Supporting], Writing—review & editing [Supporting]), Yan Jin (Conceptualization [Equal], Funding acquisition [Supporting], Supervision [Supporting], Writing—review & editing [Supporting]), and Ji Chen (Conceptualization [Equal], Funding acquisition [Lead], Supervision [Lead], Writing—review & editing [Lead]).

Conflict of interest

The authors declare no competing interests.

References

1.

Baum
A.
Stress, intrusive imagery, and chronic distress
.
Health Psychol
1990
;
9
:
653
75
.

2.

Willmore
L
,
Cameron
C
,
Yang
J
, et al. .
Behavioural and dopaminergic signatures of resilience
.
Nature
2022
;
611
:
124
32
.

3.

Woolston
C.
Stress and uncertainty drag down graduate students’ satisfaction
.
Nature
2022
;
610
:
805
8
.

4.

Haslam
C
,
Haslam
SA
,
Jetten
J
, et al. .
Life change, social identity, and health
.
Annu Rev Psychol
2021
;
72
:
635
61
.

5.

Cohen
S
,
Janicki-Deverts
D
,
Miller
GE.
Psychological stress and disease
.
JAMA
2007
;
298
:
1685
7
.

6.

Baron
KG
,
Reid
KJ.
Circadian misalignment and health
.
Int Rev Psychiatry
2014
;
26
:
139
54
.

7.

Biddie
SC
,
Conway-Campbell
BL
,
Lightman
SL.
Dynamic regulation of glucocorticoid signalling in health and disease
.
Rheumatology
2012
;
51
:
403
12
.

8.

Chrousos
GP.
Stress and disorders of the stress system
.
Nat Rev Endocrinol
2009
;
5
:
374
81
.

9.

Cruz-Pereira
JS
,
Rea
K
,
Nolan
YM
, et al. .
Depression’s unholy trinity: dysregulated stress, immunity, and the microbiome
.
Annu Rev Psychol
2020
;
71
:
49
78
.

10.

De Kloet
ER
,
Vreugdenhil
E
,
Oitzl
MS
, et al. .
Brain corticosteroid receptor balance in health and disease
.
Endocr Rev
1998
;
19
:
269
301
.

11.

Miller
GE
,
Cohen
S
,
Ritchey
AK.
Chronic psychological stress and the regulation of pro-inflammatory cytokines: a glucocorticoid-resistance model
.
Health Psychol
2002
;
21
:
531
41
.

12.

Potter
GD
,
Skene
DJ
,
Arendt
J
, et al. .
Circadian rhythm and sleep disruption: Causes, metabolic consequences, and countermeasures
.
Endocr Rev
2016
;
37
:
584
608
.

13.

Compston
JE
,
McClung
MR
,
Leslie
WD.
Osteoporosis
.
Lancet
2019
;
393
:
364
76
.

14.

Sarafrazi
N
,
Wambogo
EA
,
Shepherd
JA.
Osteoporosis or low bone mass in older adults: United States, 2017-2018
.
NCHS Data Brief
2021
;
405
:
1
8
.

15.

Ng
JS
,
Chin
KY.
Potential mechanisms linking psychological stress to bone health
.
Int J Med Sci
2021
;
18
:
604
14
.

16.

Bab
I
,
Yirmiya
R.
Depression, selective serotonin reuptake inhibitors, and osteoporosis
.
Curr Osteoporos Rep
2010
;
8
:
185
91
.

17.

Erez
HB
,
Weller
A
,
Vaisman
N
, et al. .
The relationship of depression, anxiety and stress with low bone mineral density in post-menopausal women
.
Arch Osteoporos
2012
;
7
:
247
55
.

18.

Cizza
G
,
Primma
S
,
Csako
G.
Depression as a risk factor for osteoporosis
.
Trends Endocrinol Metab
2009
;
20
:
367
73
.

19.

Pedersen
AB
,
Baggesen
LM
,
Ehrenstein
V
, et al. .
Perceived stress and risk of any osteoporotic fracture
.
Osteoporos Int
2016
;
27
:
2035
45
.

20.

El-Gabalawy
R
,
Blaney
C
,
Tsai
J
, et al. .
Physical health conditions associated with full and subthreshold PTSD in U.S. military veterans: Results from the National Health and Resilience in Veterans Study
.
J Affect Disord
2018
;
227
:
849
53
.

21.

Rauma
PH
,
Honkanen
RJ
,
Williams
LJ
, et al. .
Effects of antidepressants on postmenopausal bone loss – A 5-year longitudinal study from the OSTPRE cohort
.
Bone
2016
;
89
:
25
31
.

22.

Russell
G
,
Lightman
S.
The human stress response
.
Nat Rev Endocrinol
2019
;
15
:
525
34
.

23.

O’Connor
DB
,
Thayer
JF
,
Vedhara
K.
Stress and health: a review of psychobiological processes
.
Annu Rev Psychol
2021
;
72
:
663
88
.

24.

al’Absi
M
,
Wittmers
LE
, Jr
.
Enhanced adrenocortical responses to stress in hypertension-prone men and women
.
Ann Behav Med
2003
;
25
:
25
33
.

25.

Adam
EK
,
Quinn
ME
,
Tavernier
R
, et al. .
Diurnal cortisol slopes and mental and physical health outcomes: a systematic review and meta-analysis
.
Psychoneuroendocrinology
2017
;
83
:
25
41
.

26.

McEwen
BS.
Protective and damaging effects of stress mediators
.
N Engl J Med
1998
;
338
:
171
9
.

27.

Espinosa-Medina
I
,
Saha
O
,
Boismoreau
F
, et al. .
The sacral autonomic outflow is sympathetic
.
Science
2016
;
354
:
893
7
.

28.

Elefteriou
F.
Impact of the autonomic nervous system on the skeleton
.
Physiol Rev
2018
;
98
:
1083
112
.

29.

Tourtellotte
WG.
Axon transport and neuropathy: relevant perspectives on the etiopathogenesis of familial dysautonomia
.
Am J Pathol
2016
;
186
:
489
99
.

30.

Hasan
W.
Autonomic cardiac innervation: development and adult plasticity
.
Organogenesis
2013
;
9
:
176
93
.

31.

Goldstein
DS
,
Robertson
D
,
Esler
M
, et al. .
Dysautonomias: clinical disorders of the autonomic nervous system
.
Ann Intern Med
2002
;
137
:
753
63
.

32.

Saravia
F
,
Homo-Delarche
F.
Is innervation an early target in autoimmune diabetes?
Trends Immunol
2003
;
24
:
574
9
.

33.

Hanoun
M
,
Maryanovich
M
,
Arnal-Estape
A
, et al. .
Neural regulation of hematopoiesis, inflammation, and cancer
.
Neuron
2015
;
86
:
360
73
.

34.

Karsenty
G
,
Olson
EN.
Bone and muscle endocrine functions: unexpected paradigms of inter-organ communication
.
Cell
2016
;
164
:
1248
56
.

35.

Cappariello
A
,
Ponzetti
M
,
Rucci
N.
The “soft” side of the bone: unveiling its endocrine functions
.
Horm Mol Biol Clin Investig
2016
;
28
:
5
20
.

36.

Ducy
P
,
Amling
M
,
Takeda
S
, et al. .
Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass
.
Cell
2000
;
100
:
197
207
.

37.

Sato
S
,
Hanada
R
,
Kimura
A
, et al. .
Central control of bone remodeling by neuromedin U
.
Nat Med
2007
;
13
:
1234
40
.

38.

Togari
A.
Adrenergic regulation of bone metabolism: possible involvement of sympathetic innervation of osteoblastic and osteoclastic cells
.
Microsc Res Tech
2002
;
58
:
77
84
.

39.

Asmus
SE
,
Parsons
S
,
Landis
SC.
Developmental changes in the transmitter properties of sympathetic neurons that innervate the periosteum
.
J Neurosci
2000
;
20
:
1495
504
.

40.

Nagata
A
,
Tanaka
T
,
Minezawa
A
, et al. .
cAMP activation by PACAP/VIP stimulates IL-6 release and inhibits osteoblastic differentiation through VPAC2 receptor in osteoblastic MC3T3 cells
.
J Cell Physiol
2009
;
221
:
75
83
.

41.

Sims
NA
,
Martin
TJ.
Coupling signals between the osteoclast and osteoblast: How are messages transmitted between these temporary visitors to the bone surface?
Front Endocrinol
2015
;
6
:
41
.

42.

Rachner
TD
,
Khosla
S
,
Hofbauer
LC.
Osteoporosis: now and the future
.
Lancet
2011
;
377
:
1276
87
.

43.

Yao
Q
,
Liang
H
,
Huang
B
, et al. .
Beta-adrenergic signaling affect osteoclastogenesis via osteocytic MLO-Y4 cells’ RANKL production
.
Biochem Biophys Res Commun
2017
;
488
:
634
40
.

44.

Ma
Y
,
Nyman
JS
,
Tao
H
, et al. .
beta2-Adrenergic receptor signaling in osteoblasts contributes to the catabolic effect of glucocorticoids on bone
.
Endocrinology
2011
;
152
:
1412
22
.

45.

Kondo
A
,
Mogi
M
,
Koshihara
Y
, et al. .
Signal transduction system for interleukin-6 and interleukin-11 synthesis stimulated by epinephrine in human osteoblasts and human osteogenic sarcoma cells
.
Biochem Pharmacol
2001
;
61
:
319
26
.

46.

Yahara
M
,
Tei
K
,
Tamura
M.
Inhibition of neuropeptide Y Y1 receptor induces osteoblast differentiation in MC3T3-E1 cells
.
Mol Med Rep
2017
;
16
:
2779
84
.

47.

Wee
NKY
,
Sinder
BP
,
Novak
S
, et al. .
Skeletal phenotype of the neuropeptide Y knockout mouse
.
Neuropeptides
2019
;
73
:
78
88
.

48.

Hu
CH
,
Sui
BD
,
Liu
J
, et al. .
Sympathetic neurostress drives osteoblastic exosomal MiR-21 transfer to disrupt bone homeostasis and promote osteopenia
.
Small Methods
2022
;
6
:
e2100763
.

49.

Sui
B
,
Liu
J
,
Zheng
C
, et al. .
Targeted inhibition of osteoclastogenesis reveals the pathogenesis and therapeutics of bone loss under sympathetic neurostress
.
Int J Oral Sci
2022
;
14
:
39
.

50.

Elefteriou
F
,
Ahn
JD
,
Takeda
S
, et al. .
Leptin regulation of bone resorption by the sympathetic nervous system and CART
.
Nature
2005
;
434
:
514
20
.

51.

Holmes
TH
,
Rahe
RH.
The social readjustment rating scale
.
J Psychosom Res
1967
;
11
:
213
8
.

52.

McEwen
BS.
Physiology and neurobiology of stress and adaptation: central role of the brain
.
Physiol Rev
2007
;
87
:
873
904
.

53.

Wirtz
PH
,
von Kanel
R.
Psychological stress, inflammation, and coronary heart disease
.
Curr Cardiol Rep
2017
;
19
:
111
.

54.

Antelman
SM
,
DeGiovanni
LA
,
Kocan
D.
A single exposure to cocaine or immobilization stress provides extremely long-lasting, selective protection against sudden cardiac death from tetracaine
.
Life Sci
1989
;
44
:
201
7
.

55.

Van Dijken
HH
,
Van der Heyden
JA
,
Mos
J
, et al. .
Inescapable footshocks induce progressive and long-lasting behavioural changes in male rats
.
Physiol Behav
1992
;
51
:
787
94
.

56.

Pynoos
RS
,
Ritzmann
RF
,
Steinberg
AM
, et al. .
A behavioral animal model of posttraumatic stress disorder featuring repeated exposure to situational reminders
.
Biol Psychiatry
1996
;
39
:
129
34
.

57.

Siegmund
A
,
Wotjak
CT.
A mouse model of posttraumatic stress disorder that distinguishes between conditioned and sensitised fear
.
J Psychiatr Res
2007
;
41
:
848
60
.

58.

Morris
R.
Developments of a water-maze procedure for studying spatial learning in the rat
.
J Neurosci Methods
1984
;
11
:
47
60
.

59.

Richter-Levin
G.
Acute and long-term behavioral correlates of underwater trauma--potential relevance to stress and post-stress syndromes
.
Psychiatry Res
1998
;
79
:
73
83
.

60.

Balkaya
M
,
Prinz
V
,
Custodis
F
, et al. .
Stress worsens endothelial function and ischemic stroke via glucocorticoids
.
Stroke
2011
;
42
:
3258
64
.

61.

Gameiro
GH
,
Gameiro
PH
,
Andrade Ada
S
, et al. .
Nociception- and anxiety-like behavior in rats submitted to different periods of restraint stress
.
Physiol Behav
2006
;
87
:
643
9
.

62.

Nakajima
K
,
Hamada
N
,
Takahashi
Y
, et al. .
Restraint stress enhances alveolar bone loss in an experimental rat model
.
J Periodontal Res
2006
;
41
:
527
34
.

63.

Takeda
O
,
Toyama
T
,
Watanabe
K
, et al. .
Ameliorating effects of Juzentaihoto on restraint stress and P. gingivalis-induced alveolar bone loss
.
Arch Oral Biol
2014
;
59
:
1130
8
.

64.

Liberzon
I
,
Krstov
M
,
Young
EA.
Stress-restress: effects on ACTH and fast feedback
.
Psychoneuroendocrinology
1997
;
22
:
443
53
.

65.

Adamec
RE
,
Shallow
T.
Lasting effects on rodent anxiety of a single exposure to a cat
.
Physiol Behav
1993
;
54
:
101
9
.

66.

Zoladz
PR
,
Conrad
CD
,
Fleshner
M
, et al. .
Acute episodes of predator exposure in conjunction with chronic social instability as an animal model of post-traumatic stress disorder
.
Stress
2008
;
11
:
259
81
.

67.

Zhang
J
,
Fujita
Y
,
Chang
L
, et al. .
Beneficial effects of anti-RANKL antibody in depression-like phenotype, inflammatory bone markers, and bone mineral density in male susceptible mice after chronic social defeat stress
.
Behav Brain Res
2020
;
379
:
112397
.

68.

Hammamieh
R
,
Chakraborty
N
,
De Lima
TC
, et al. .
Murine model of repeated exposures to conspecific trained aggressors simulates features of post-traumatic stress disorder
.
Behav Brain Res
2012
;
235
:
55
66
.

69.

Pibiri
F
,
Nelson
M
,
Guidotti
A
, et al. .
Decreased corticolimbic allopregnanolone expression during social isolation enhances contextual fear: a model relevant for posttraumatic stress disorder
.
Proc Natl Acad Sci U S A
2008
;
105
:
5567
72
.

70.

Mountain
RV
,
Langlais
AL
,
Hu
D
, et al. .
Social isolation through single housing negatively affects trabecular and cortical bone in adult male, but not female, C57BL/6J mice
.
Bone
2023
;
172
:
116762
.

71.

Patki
G
,
Solanki
N
,
Salim
S.
Witnessing traumatic events causes severe behavioral impairments in rats
.
Int J Neuropsychopharmacol
2014
;
17
:
2017
29
.

72.

Ardi
Z
,
Albrecht
A
,
Richter-Levin
A
, et al. .
Behavioral profiling as a translational approach in an animal model of posttraumatic stress disorder
.
Neurobiol Dis
2016
;
88
:
139
47
.

73.

Kalinichev
M
,
Easterling
KW
,
Plotsky
PM
, et al. .
Long-lasting changes in stress-induced corticosterone response and anxiety-like behaviors as a consequence of neonatal maternal separation in Long-Evans rats
.
Pharmacol Biochem Behav
2002
;
73
:
131
40
.

74.

Bertolini Botelho
MC
,
Cintra
LTA
,
da Silva
CC
, et al. .
Early life stress exacerbates bone resorption and inhibits anxiety-like behaviour induced by apical periodontitis in rats
.
Int Endod J
2023
;
56
:
203
12
.

75.

Steru
L
,
Chermat
R
,
Thierry
B
, et al. .
The tail suspension test: a new method for screening antidepressants in mice
.
Psychopharmacology
1985
;
85
:
367
70
.

76.

Hu
Y
,
Zhang
Y
,
Ni
CY
, et al. .
Human umbilical cord mesenchymal stromal cells-derived extracellular vesicles exert potent bone protective effects by CLEC11A-mediated regulation of bone metabolism
.
Theranostics
2020
;
10
:
2293
308
.

77.

Reber
SO
,
Birkeneder
L
,
Veenema
AH
, et al. .
Adrenal insufficiency and colonic inflammation after a novel chronic psycho-social stress paradigm in mice: implications and mechanisms
.
Endocrinology
2007
;
148
:
670
82
.

78.

Xie
X
,
Shen
Q
,
Yu
C
, et al. .
Depression-like behaviors are accompanied by disrupted mitochondrial energy metabolism in chronic corticosterone-induced mice
.
J Steroid Biochem Mol Biol
2020
;
200
:
105607
.

79.

Fetcho
RN
,
Hall
BS
,
Estrin
DJ
, et al. .
Regulation of social interaction in mice by a frontostriatal circuit modulated by established hierarchical relationships
.
Nat Commun
2023
;
14
:
2487
.

80.

Francois
M
,
Canal Delgado
I
,
Shargorodsky
N
, et al. .
Assessing the effects of stress on feeding behaviors in laboratory mice
.
Elife
2022
;
11
:
e70271
.

81.

Lee
EH
,
Park
JY
,
Kwon
HJ
, et al. .
Repeated exposure with short-term behavioral stress resolves pre-existing stress-induced depressive-like behavior in mice
.
Nat Commun
2021
;
12
:
6682
.

82.

Liu
MY
,
Yin
CY
,
Zhu
LJ
, et al. .
Sucrose preference test for measurement of stress-induced anhedonia in mice
.
Nat Protoc
2018
;
13
:
1686
98
.

83.

Shi
X
,
Gao
Y
,
Song
L
, et al. .
Sulfur dioxide derivatives produce antidepressant- and anxiolytic-like effects in mice
.
Neuropharmacology
2020
;
176
:
108252
.

84.

Sui
B
,
Hu
C
,
Liao
L
, et al. .
Mesenchymal progenitors in osteopenias of diverse pathologies: differential characteristics in the common shift from osteoblastogenesis to adipogenesis
.
Sci Rep
2016
;
6
:
30186
.

85.

Young
BE
,
Holwerda
SW
,
Vranish
JR
, et al. .
Sympathetic transduction in type 2 diabetes mellitus
.
Hypertension
2019
;
74
:
201
7
.

86.

Rana
T
,
Schultz
MA
,
Freeman
ML
, et al. .
Loss of Nrf2 accelerates ionizing radiation-induced bone loss by upregulating RANKL
.
Free Radic Biol Med
2012
;
53
:
2298
307
.

87.

Filipowska
J
,
Tomaszewski
KA
,
Niedzwiedzki
L
, et al. .
The role of vasculature in bone development, regeneration and proper systemic functioning
.
Angiogenesis
2017
;
20
:
291
302
.

88.

Maes
C
,
Carmeliet
G
,
Schipani
E.
Hypoxia-driven pathways in bone development, regeneration and disease
.
Nat Rev Rheumatol
2012
;
8
:
358
66
.

89.

Clarkin
C
,
Olsen
BR.
On bone-forming cells and blood vessels in bone development
.
Cell Metab
2010
;
12
:
314
6
.

90.

Duan
X
,
Murata
Y
,
Liu
Y
, et al. .
Vegfa regulates perichondrial vascularity and osteoblast differentiation in bone development
.
Development
2015
;
142
:
1984
91
.

91.

Kusumbe
AP
,
Ramasamy
SK
,
Adams
RH.
Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone
.
Nature
2014
;
507
:
323
8
.

92.

Nakashima
K
,
Zhou
X
,
Kunkel
G
, et al. .
The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation
.
Cell
2002
;
108
:
17
29
.

93.

Langen
UH
,
Pitulescu
ME
,
Kim
JM
, et al. .
Cell-matrix signals specify bone endothelial cells during developmental osteogenesis
.
Nat Cell Biol
2017
;
19
:
189
201
.

94.

Watson
EC
,
Adams
RH.
Biology of bone: the vasculature of the skeletal system
.
Cold Spring Harb Perspect Med
2018
;
8
:
a031559
.

95.

Burnstock
G.
Local mechanisms of blood flow control by perivascular nerves and endothelium
.
J Hypertens Suppl
1990
;
8
:
S95
106
.

96.

Godo
S
,
Shimokawa
H.
Endothelial functions
.
Arterioscler Thromb Vasc Biol
2017
;
37
:
e108
14
.

97.

Tesfamariam
B
,
Weisbrod
RM
,
Cohen
RA.
Endothelium inhibits responses of rabbit carotid artery to adrenergic nerve stimulation
.
Am J Physiol
1987
;
253
:
H792
798
.

98.

Guimaraes
S
,
Moura
D.
Vascular adrenoceptors: an update
.
Pharmacol Rev
2001
;
53
:
319
56
.

99.

Luscher
TF
,
Yang
Z
,
Tschudi
M
, et al. .
Interaction between endothelin-1 and endothelium-derived relaxing factor in human arteries and veins
.
Circ Res
1990
;
66
:
1088
94
.

100.

Potente
M
,
Gerhardt
H
,
Carmeliet
P.
Basic and therapeutic aspects of angiogenesis
.
Cell
2011
;
146
:
873
87
.

101.

Makanya
AN
,
Hlushchuk
R
,
Djonov
VG.
Intussusceptive angiogenesis and its role in vascular morphogenesis, patterning, and remodeling
.
Angiogenesis
2009
;
12
:
113
23
.

102.

Djonov
V
,
Baum
O
,
Burri
PH.
Vascular remodeling by intussusceptive angiogenesis
.
Cell Tissue Res
2003
;
314
:
107
17
.

103.

Blanco
R
,
Gerhardt
H.
VEGF and Notch in tip and stalk cell selection
.
Cold Spring Harb Perspect Med
2013
;
3
:
a006569
.

104.

Gerhardt
H
,
Golding
M
,
Fruttiger
M
, et al. .
VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia
.
J Cell Biol
2003
;
161
:
1163
77
.

105.

Tymko
MM
,
Lawley
JS
,
Ainslie
PN
, et al. .
Global Reach 2018 Heightened alpha-Adrenergic Signaling Impairs Endothelial Function During Chronic Exposure to Hypobaric Hypoxia
.
Circ Res
2020
;
127
:
e1
e13
.

106.

Swenson
ER.
Sympathetic nervous system activation and vascular endothelial function with chronic hypoxia
.
Circ Res
2020
;
127
:
247
8
.

107.

Ungvari
Z
,
Tarantini
S
,
Kiss
T
, et al. .
Endothelial dysfunction and angiogenesis impairment in the ageing vasculature
.
Nat Rev Cardiol
2018
;
15
:
555
65
.

108.

Chen
HI
,
Li
HT
,
Chen
CC.
Physical conditioning decreases norepinephrine-induced vasoconstriction in rabbits. Possible roles of norepinephrine-evoked endothelium-derived relaxing factor
.
Circulation
1994
;
90
:
970
5
.

109.

Kalluri
R
,
LeBleu
VS.
The biology, function, and biomedical applications of exosomes
.
Science
2020
;
367
:
eaau6977
.

110.

Harding
C
,
Heuser
J
,
Stahl
P.
Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes
.
J Cell Biol
1983
;
97
:
329
39
.

111.

Pan
BT
,
Teng
K
,
Wu
C
, et al. .
Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes
.
J Cell Biol
1985
;
101
:
942
8
.

112.

Raposo
G
,
Nijman
HW
,
Stoorvogel
W
, et al. .
B lymphocytes secrete antigen-presenting vesicles
.
J Exp Med
1996
;
183
:
1161
72
.

113.

Huang-Doran
I
,
Zhang
CY
,
Vidal-Puig
A
, et al.
Extracellular vesicles: novel mediators of cell communication in metabolic disease
.
Trends Endocrinol Metab
2017
;
28
:
3
18
.

114.

Gurunathan
S
,
Kang
MH
,
Jeyaraj
M
, et al. .
Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes
.
Cells
2019
;
8
:
307
.

115.

Takahashi
A
,
Okada
R
,
Nagao
K
, et al. .
Exosomes maintain cellular homeostasis by excreting harmful DNA from cells
.
Nat Commun
2017
;
8
:
15287
.

116.

Mathieu
M
,
Martin-Jaular
L
,
Lavieu
G
, et al. .
Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication
.
Nat Cell Biol
2019
;
21
:
9
17
.

117.

Thery
C
,
Zitvogel
L
,
Amigorena
S.
Exosomes: composition, biogenesis and function
.
Nat Rev Immunol
2002
;
2
:
569
79
.

118.

Pegtel
DM
,
Gould
SJ.
Exosomes
.
Annu Rev Biochem
2019
;
88
:
487
514
.

119.

Liu
J
,
Ren
L
,
Li
S
, et al. .
The biology, function, and applications of exosomes in cancer
.
Acta Pharm Sin B
2021
;
11
:
2783
97
.

120.

van Niel
G
,
D’Angelo
G
,
Raposo
G.
Shedding light on the cell biology of extracellular vesicles
.
Nat Rev Mol Cell Biol
2018
;
19
:
213
28
.

121.

McAndrews
KM
,
Kalluri
R.
Mechanisms associated with biogenesis of exosomes in cancer
.
Mol Cancer
2019
;
18
:
52
.

122.

Thery
C
,
Witwer
KW
,
Aikawa
E
, et al. .
Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines
.
J Extracell Vesicles
2018
;
7
:
1535750
.

123.

Shah
R
,
Patel
T
,
Freedman
JE.
Circulating extracellular vesicles in human disease
.
N Engl J Med
2018
;
379
:
958
66
.

124.

Alvarez-Erviti
L
,
Seow
Y
,
Yin
H
, et al. .
Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes
.
Nat Biotechnol
2011
;
29
:
341
5
.

125.

Kamerkar
S
,
LeBleu
VS
,
Sugimoto
H
, et al. .
Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer
.
Nature
2017
;
546
:
498
503
.

126.

Wang
Q
,
Yu
J
,
Kadungure
T
, et al. .
ARMMs as a versatile platform for intracellular delivery of macromolecules
.
Nat Commun
2018
;
9
:
960
.

127.

Taraboletti
G
,
D’Ascenzo
S
,
Borsotti
P
, et al. .
Shedding of the matrix metalloproteinases MMP-2, MMP-9, and MT1-MMP as membrane vesicle-associated components by endothelial cells
.
Am J Pathol
2002
;
160
:
673
80
.

128.

Critser
PJ
,
Yoder
MC.
Endothelial colony-forming cell role in neoangiogenesis and tissue repair
.
Curr Opin Organ Transplant
2010
;
15
:
68
72
.

129.

Deregibus
MC
,
Cantaluppi
V
,
Calogero
R
, et al. .
Endothelial progenitor cell derived microvesicles activate an angiogenic program in endothelial cells by a horizontal transfer of mRNA
.
Blood
2007
;
110
:
2440
8
.

130.

Giricz
Z
,
Varga
ZV
,
Baranyai
T
, et al. .
Cardioprotection by remote ischemic preconditioning of the rat heart is mediated by extracellular vesicles
.
J Mol Cell Cardiol
2014
;
68
:
75
8
.

131.

Femmino
S
,
Penna
C
,
Margarita
S
, et al. .
Extracellular vesicles and cardiovascular system: biomarkers and Cardioprotective Effectors
.
Vascul Pharmacol
2020
;
135
:
106790
.

132.

Potz
BA
,
Scrimgeour
LA
,
Pavlov
VI
, et al. .
Extracellular vesicle injection improves myocardial function and increases angiogenesis in a swine model of chronic ischemia
.
J Am Heart Assoc
2018
;
7
:
e008344
.

133.

Cantaluppi
V
,
Gatti
S
,
Medica
D
, et al. .
Microvesicles derived from endothelial progenitor cells protect the kidney from ischemia-reperfusion injury by microRNA-dependent reprogramming of resident renal cells
.
Kidney Int
2012
;
82
:
412
27
.

134.

Zhang
J
,
Chen
C
,
Hu
B
, et al. .
Exosomes derived from human endothelial progenitor cells accelerate cutaneous wound healing by promoting angiogenesis through Erk1/2 signaling
.
Int J Biol Sci
2016
;
12
:
1472
87
.

135.

Zhuang
Y
,
Cheng
M
,
Li
M
, et al. .
Small extracellular vesicles derived from hypoxic mesenchymal stem cells promote vascularized bone regeneration through the miR-210-3p/EFNA3/PI3K pathway
.
Acta Biomater
2022
;
150
:
413
26
.

136.

Liu
L
,
Zheng
CX
,
Zhao
N
, et al. .
Mesenchymal stem cell aggregation-released extracellular vesicles induce CD31(+) EMCN(+) vessels in skin regeneration and improve diabetic wound healing
.
Adv Healthc Mater
2023
;
12
:
e2300019
.

137.

Friedenstein
AJ
,
Chailakhyan
RK
,
Gerasimov
UV.
Bone marrow osteogenic stem cells: in vitro cultivation and transplantation in diffusion chambers
.
Cell Tissue Kinet
1987
;
20
:
263
72
.

138.

Owen
M
,
Friedenstein
AJ.
Stromal stem cells: marrow-derived osteogenic precursors
.
Ciba Found Symp
1988
;
136
:
42
60
.

139.

Samsonraj
RM
,
Raghunath
M
,
Nurcombe
V
, et al. .
Concise review: multifaceted characterization of human mesenchymal stem cells for use in regenerative medicine
.
Stem Cells Transl Med
2017
;
6
:
2173
85
.

140.

Timmers
L
,
Lim
SK
,
Arslan
F
, et al. .
Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium
.
Stem Cell Res
2007
;
1
:
129
37
.

141.

van Koppen
A
,
Joles
JA
,
van Balkom
BW
, et al. .
Human embryonic mesenchymal stem cell-derived conditioned medium rescues kidney function in rats with established chronic kidney disease
.
PLoS One
2012
;
7
:
e38746
.

142.

Lai
RC
,
Arslan
F
,
Lee
MM
, et al. .
Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury
.
Stem Cell Res
2010
;
4
:
214
22
.

143.

Ono
M
,
Kosaka
N
,
Tominaga
N
, et al. .
Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells
.
Sci Signaling
2014
;
7
:
ra63
.

144.

Phinney
DG
,
Di Giuseppe
M
,
Njah
J
, et al. .
Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs
.
Nat Commun
2015
;
6
:
8472
.

145.

Rani
S
,
Ryan
AE
,
Griffin
MD
, et al. .
Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications
.
Mol Ther
2015
;
23
:
812
23
.

146.

Eirin
A
,
Zhu
XY
,
Puranik
AS
, et al. .
Mesenchymal stem cell-derived extracellular vesicles attenuate kidney inflammation
.
Kidney Int
2017
;
92
:
114
24
.

147.

Haga
H
,
Yan
IK
,
Takahashi
K
, et al. .
Extracellular vesicles from bone marrow-derived mesenchymal stem cells improve survival from lethal hepatic failure in mice
.
Stem Cells Transl Med
2017
;
6
:
1262
72
.

148.

Li
L
,
Zhang
Y
,
Mu
J
, et al. .
Transplantation of human mesenchymal stem-cell-derived exosomes immobilized in an adhesive hydrogel for effective treatment of spinal cord injury
.
Nano Lett
2020
;
20
:
4298
305
.

149.

Liu
W
,
Li
L
,
Rong
Y
, et al. .
Hypoxic mesenchymal stem cell-derived exosomes promote bone fracture healing by the transfer of miR-126
.
Acta Biomater
2020
;
103
:
196
212
.

150.

Tao
SC
,
Yuan
T
,
Zhang
YL
, et al. .
Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model
.
Theranostics
2017
;
7
:
180
95
.

151.

Kishore
R
,
Khan
M.
More than tiny sacks: stem cell exosomes as cell-free modality for cardiac repair
.
Circ Res
2016
;
118
:
330
43
.

152.

Haney
MJ
,
Klyachko
NL
,
Zhao
Y
, et al. .
Exosomes as drug delivery vehicles for Parkinson’s disease therapy
.
J Control Release
2015
;
207
:
18
30
.

153.

Yu
M
,
Liu
W
,
Li
J
, et al. .
Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway
.
Stem Cell Res Ther
2020
;
11
:
350
.

154.

Fatima
F
,
Ekstrom
K
,
Nazarenko
I
, et al. .
Non-coding RNAs in mesenchymal stem cell-derived extracellular vesicles: deciphering regulatory roles in stem cell potency, inflammatory resolve, and tissue regeneration
.
Front Genet
2017
;
8
:
161
.

155.

Teng
X
,
Chen
L
,
Chen
W
, et al. .
Mesenchymal stem cell-derived exosomes improve the microenvironment of infarcted myocardium contributing to angiogenesis and anti-inflammation
.
Cell Physiol Biochem
2015
;
37
:
2415
24
.

156.

Zhang
HC
,
Liu
XB
,
Huang
S
, et al. .
Microvesicles derived from human umbilical cord mesenchymal stem cells stimulated by hypoxia promote angiogenesis both in vitro and in vivo
.
Stem Cells Dev
2012
;
21
:
3289
97
.

157.

Zou
X
,
Gu
D
,
Xing
X
, et al. .
Human mesenchymal stromal cell-derived extracellular vesicles alleviate renal ischemic reperfusion injury and enhance angiogenesis in rats
.
Am J Transl Res
2016
;
8
:
4289
99
.

158.

Ma
J
,
Zhao
Y
,
Sun
L
, et al. .
Exosomes Derived from Akt-Modified Human Umbilical Cord Mesenchymal Stem Cells Improve Cardiac Regeneration and Promote Angiogenesis via Activating Platelet-Derived Growth Factor D
.
Stem Cells Transl Med
2017
;
6
:
51
9
.

159.

Wu
M
,
Liu
X
,
Li
Z
, et al. .
SHED aggregate exosomes shuttled miR-26a promote angiogenesis in pulp regeneration via TGF-beta/SMAD2/3 signalling
.
Cell Prolif
2021
;
54
:
e13074
.

160.

Chen
S
,
Tang
Y
,
Liu
Y
, et al. .
Exosomes derived from miR-375-overexpressing human adipose mesenchymal stem cells promote bone regeneration
.
Cell Prolif
2019
;
52
:
e12669
.

161.

Li
W
,
Liu
Y
,
Zhang
P
, et al. .
Tissue-engineered bone immobilized with human adipose stem cells-derived exosomes promotes bone regeneration
.
ACS Appl Mater Interfaces
2018
;
10
:
5240
54
.

162.

Li
X
,
Zheng
Y
,
Hou
L
, et al. .
Exosomes derived from maxillary BMSCs enhanced the osteogenesis in iliac BMSCs
.
Oral Dis
2020
;
26
:
131
44
.

163.

Qi
X
,
Zhang
J
,
Yuan
H
, et al. .
Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells repair critical-sized bone defects through enhanced angiogenesis and Osteogenesis in Osteoporotic rats
.
Int J Biol Sci
2016
;
12
:
836
49
.

164.

Liao
W
,
Ning
Y
,
Xu
HJ
, et al. .
BMSC-derived exosomes carrying microRNA-122-5p promote proliferation of osteoblasts in osteonecrosis of the femoral head
.
Clin Sci
2019
;
133
:
1955
75
.

165.

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

166.

Zhang
J
,
Liu
X
,
Li
H
, et al. .
Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signaling pathway
.
Stem Cell Res Ther
2016
;
7
:
136
.

167.

Lu
Z
,
Chen
Y
,
Dunstan
C
, et al. .
Priming adipose stem cells with tumor necrosis factor-alpha preconditioning potentiates their exosome efficacy for bone regeneration
.
Tissue Eng Part A
2017
;
23
:
1212
20
.

168.

Zhu
Q
,
Heon
M
,
Zhao
Z
, et al. .
Microfluidic engineering of exosomes: editing cellular messages for precision therapeutics
.
Lab Chip
2018
;
18
:
1690
703
.

169.

Sun
D
,
Zhuang
X
,
Xiang
X
, et al. .
A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes
.
Mol Ther
2010
;
18
:
1606
14
.

170.

Pascucci
L
,
Cocce
V
,
Bonomi
A
, et al. .
Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery
.
J Control Release
2014
;
192
:
262
70
.

171.

Kim
MS
,
Haney
MJ
,
Zhao
Y
, et al. .
Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells
.
Nanomedicine
2016
;
12
:
655
64
.

172.

Fuhrmann
G
,
Serio
A
,
Mazo
M
, et al. .
Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins
.
J Control Release
2015
;
205
:
35
44
.

173.

Sato
YT
,
Umezaki
K
,
Sawada
S
, et al. .
Engineering hybrid exosomes by membrane fusion with liposomes
.
Sci Rep
2016
;
6
:
21933
.

Author notes

Hao-Kun Xu, Jie-Xi Liu and Ze-Kai Zhou contributed equally to this work.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.