Your privacy, your choice

We use essential cookies to make sure the site can function. We also use optional cookies for advertising, personalisation of content, usage analysis, and social media.

By accepting optional cookies, you consent to the processing of your personal data - including transfers to third parties. Some third parties are outside of the European Economic Area, with varying standards of data protection.

See our privacy policy for more information on the use of your personal data.

for further information and to change your choices.

You are viewing the site in preview mode

Skip to main content

Cellular crosstalk in the bone marrow niche

Abstract

The bone marrow niche is a special microenvironment that comprises elements, including hematopoietic stem cells, osteoblasts, and endothelial cells, and helps maintain their characteristic functions. Here, we elaborate on the crosstalk between various cellular components, hematopoietic stem cells, and other cells in the bone marrow niche. We further explain the mechanism of preserving equilibrium in the bone marrow niche, which is crucial for the directional regulation of bone reconstruction and repair. Additionally, we elucidate the intercommunication among osteocytes, the regulation of osteoblast maturation and activation by lymphocytes, the deficiency of megakaryocytes that can markedly impair osteoblast formation, and the mechanism of interaction between macrophages and mesenchymal stem cells in the bone marrow niche. Finally, we discussed the new immunotherapies for bone tumors in the BM niche. In this review, we aimed to provide a candid overview of the crosstalk among bone marrow niche cells and to highlight new concepts underlying the unknown mechanisms of hematopoiesis and bone reconstruction. Thus, this review may provide a more comprehensive understanding of the role of these niche cells in improving hematopoietic function and help identify their therapeutic potential for different diseases in the future.

Graphical Abstract

Introduction

Osteoporosis is a representative disease of bone remodeling imbalance caused by aging or disease status [1]. Currently, no effective treatment exists to reverse bone metabolism imbalance and induce the directional reconstruction and repair of bone [2]. Nevertheless, the precise elucidation and analysis of cellular activities during bone remodeling and repair, improvement of hematopoietic function, and restoration of bone formation will greatly aid in the treatment of bone diseases and bone degradation in the rapidly increasing aging population in China and even in the global population.

The bone marrow (BM) niche is a special microenvironment that is composed of hematopoietic stem cells (HSCs), osteoblasts, adipocytes, and endothelial cells and helps maintain the unique functions of the BM [3]. The BM niche ensures hematopoietic homeostasis by controlling the proliferation, self-renewal, differentiation, and migration of HSCs and progenitor cells, as well as the responses to emergencies and injuries [4]. This niche comprises a unique matrix microenvironment required to support the hematopoietic function of HSCs [4]. The BM niche cells exert their effects on HSCs via three pathways: intercellular communication, cell adhesion, and extracellular matrix (ECM) remodeling [5]. Among the extrinsic factors released by these HSC-supporting cells, the chemokine stromal cell-derived factor 1 [SDF-1, also known as chemokine [C-X-C motif] ligand 12 [CXCL12]) and the cytokine stem cell factor (SCF) are essential regulators that manage HSCs in the BM [6]. Over the last decade, remarkable progress has been achieved in deciphering the BM niche and the specific cellular and molecular elements that regulate HSC activity, identifying the major cellular components within the BM niche, and determining the mechanisms of cytokine-mediated cellular interaction. However, our understanding of the BM niche is still in its early stages. Therefore, in this review, we summarize the cellular components and intercellular crosstalk in the BM niche to provide a basis for the further investigation of the role of the BM niche in the maintenance of hematopoietic homeostasis and facilitate subsequent research on the targeted treatment of diseases.

Role of bone marrow niche in maintaining the balance of bone remodeling

In modern mammals, the BM consists of many different cell types, ECM, secreted factors (such as cytokines, chemokines, and growth factors), and metabolic signals [7] (Fig. 1). The HSCs in the BM promote bone remodeling/repair by dynamically interacting with a complex network of stromal cells, including by the reciprocal molecular crosstalk among inflammatory, endothelial, and niche cells, such as mesenchymal stem cells and schwann cells [8, 9]. Furthermore, osteoblast function during bone repair relies on signals from the surrounding niche cells, which provide a specific microenvironment and integrate signals to mediate appropriate stem cell responses, including osteogenic/chondrogenic differentiation, depending on the need of the organism [10]. Osteogenesis is related to the H-type endothelial cell subtype of capillary endothelial cells [10]. H-type cells express platelet and endothelial cell adhesion molecule 1 (PECAM1) and intrinsic mucin, targeting the molecular signals of bone progenitor cells through the Notch signaling pathway, reducing the expression of secreted phosphoprotein 1 (Spp1), RUNX family transcription factor 2 (Runx2), and Osterix (Osx), changing the BM niche of the injury site, and causing bone progenitor cells to differentiate into osteoblasts [11]. Inflammatory macrophages play an important role in endochondral ossification during fracture healing [10]. The influx of inflammatory cells leads to the secretion of bone morphogenetic protein 4 (BMP4), vascular endothelial growth factor (VEGF), interleukin 6 (IL6), and C–C motif chemokine ligand 2 (CCL2), which promote osteogenic differentiation and bone repair by interacting with newly formed bone progenitor cells [12]. In addition, sensory and sympathetic nerves play a crucial role in bone homeostasis and bone repair [10]. Nerve growth factor (NGF) expressed by nerve cells and VEGF expressed by Schwann cells are upregulated during bone repair [13].

Fig. 1
figure 1

Components of the HSPC bone marrow microenvironment. The bone marrow cavity contains multiple cell types from different lineages, and hematopoietic stem cells (HSCs) maintain hematopoiesis. Hematopoietic stem cells are predominantly found in perivascular niches (including periarterial, perisinusoidal, and perivascular in the transition zone). Osteoblastic niches are located near the periosteum, where they communicate locally through cell–cell interactions and remotely through cytokines or exosomes released into the bone marrow. HSC, hematopoietic stem cell; CAR, CXCL12-rich reticulocyte; ECM, extracellular matrix

Overall, the dynamic equilibrium of the bone microenvironment regulates the regeneration and repair of bone, with the BM being the main site for the balance between bone regeneration and hematopoiesis. Thus, the bone microenvironment is important in the control and maintenance of the HSC niche. Table 1 summarizes the various cells of the bone microenvironment and their main markers in the HSC niche, as well as the main molecular crosstalk effects and cellular functions with HSCs (Table 1). Moreover, the interaction of endothelial cells and osteoblasts and the niche function are essential for maintaining bone health and coordinating bone repair and regeneration [14]. In addition, BM macrophages, megakaryocytes (MKs), CXCL12-abundant reticular (CAR) cells, and other cells that constitute the cellular components of the BM niche are vital in maintaining the functional balance of the niche [14, 15]. Aged skeletal stem cells can create an inflammatory degenerative niche [16]. In bone, cell-to-cell interactions maintain the dynamic microenvironment of the bone, wherein the crosstalk between osteoblast and vascular lineage cells and inflammatory cells is particularly essential for bone repair and remodeling [17]. Furthermore, the niche interacts with bone cells during homeostasis to preserve their undifferentiated properties. However, the changing niche environment after bone injury causes precursor cells to differentiate into osteoblasts and repair the damaged tissue [10].

Table 1 Different cell types, markers and functions within the niche

Cellular crosstalk in the bone marrow niche

Interaction of hematopoietic stem cells with other cells in their niche

Mesenchymal stem cells and hematopoietic stem cells

BM mesenchymal stem cells (MSCs) are a group of cells that perform multiple functions and are located around blood vessels as well as directly on blood vessel surfaces along with sympathetic nerves [17]. The origins of MSC development are still being investigated, wherein MSCs have been initially thought to be derived from the mesoderm [18]. Furthermore, studies have shown that MSCs with positive (CD73, CD90, and CD105) and negative (CD14, CD19, CD34, CD45, and HLA-DR) markers can differentiate into other cell types, such as osteoblasts, chondrocytes, neural cells, muscle cells, and adipocytes, as well as secrete various growth factors, cytokines, and chemokines [19, 20]. Commonly used MSCs include BM-derived MSCs, fat-derived MSCs, and umbilical cord blood-derived MSCs [21]. Moreover, the substances secreted by MSCs are important in immune regulation, cell migration, proliferation, differentiation, and tissue repair.

Several MSC populations, including Nes-GFP+, NG2+, LEPR+ MSCs, or CAR cells, promote the maintenance of HSCs by releasing factors such as CXCL12 or SCF [19, 20]. MSCs are one of the main elements that promote the self-renewal niche of HSCs, where MSCs in the BM and cord blood express high levels of N-cadherin (NCAD), vascular cell adhesion molecule 1 (VCAM-1), neural cell adhesion molecule 1 (NCAM-1), and integrins [22]. Moreover, the interaction between MSCs and HSCs is multifaceted and plays a prominent role in supporting HSCs, including the transmission of the Wnt and Notch signaling pathways [23]. This balance between the HSCs and MSCs guarantees self-renewal and maintenance of the HSC niche. BMP4, a member of another signaling pathway secreted by MSC, influences HSCs during embryonic development, within which the conditional inactivation of the BMP4 receptor results in an increased HSC population in the BM [24]. Additionally, BMP4 can affect HSCs directly or through mediators such as Shh (Sonic hedgehog), in which Shh induces cytokine-dependent proliferation of HSCs, e.g., high concentrations of the Shh protein support the proliferation of cord blood HSCs [25]. The platelet derived growth factor receptor (PDGFR)-α+ CD51+ subpopulation of nestin (Nes)+ cells have also been found to be rich in major HSC maintenance genes, supporting the notion of the co-separation of niche activity from MSC activity [26]. These findings suggest that cell-to-cell contact is one of the main mechanisms for maintaining HSC self-renewal by MSCs.

CAR cells and hematopoietic stem cells

CAR cells, also known as leptin receptor (LEPR)-expressing cells or CXCL12- and high SCF-expressing reticular cells, are a population of MSCs adjacent to blood sinuses [6]. The CAR cell subset is the main cellular component of the hematopoietic stem and progenitor cell (HSPC), creating a niche for HSCs capable of regulating HSC self-renewal, proliferation, and migration [27] (Fig. 2). CAR cells were the first cells found to colocalize with HSCs in the BM [28]. Although CAR/LEPR+ MSCs constitute approximately 0.3% of the BM cells (including almost all colony-forming unit [CFU]-fibroblasts), they represent the major source of adipocytes and osteoblasts in the BM of adult mice and are required for HSC maintenance [28]. However, the CAR-expressing cell types are not well defined, with studies suggesting that these cells express genes including nestin, anti-myxovirus-1, LEPR, transcription factor damage-related homeobox (Prx-1), and osteogenesis-related transcription factor osterix (OSX) [29]. CAR cells, osteoblasts, and MSCs are niche components closely related to HSCs and perform important functions in immune cell function and hematopoiesis homeostasis [30]. Furthermore, CAR cells inversely regulate HSCs to create a niche and simultaneously generate osteoblasts to ensure sufficient physical space for HSCs [30].

Fig. 2
figure 2

Model showing the CAR cells, comprising the bone marrow hematopoietic stem cell (HSC) niche. CAR cells rich in LEPR+, Nes-GFP+, and CXCL12-GFP+in the sinusoids secrete SCF to maintain HSCs in the niche and CXCL12 to achieve self-renewal of HSCs, while MSCs containing NG2+and Nes-GFP+ in arteriole vessels can only secrete CXCL12 to achieve self-renewal of HSCs

Interestingly, CAR cells with high expression of CXCL12 and SCF make up the majority of the perisinusoidal niche, and deleting CXCL12 or SCF from all MSCs, in particular, leads to the depletion of HSC populations [6]. Moreover, CAR cells highly express lipid and osteogenic factors, such as peroxisome proliferator-activated receptor γ (PPAR-γ) and Runx, and can differentiate into osteoblasts and adipocytes [28, 31]. Studies have reported that mouse CAR cells undergo short-term ablation when CXCL12 and SCF production decreases, resulting in decreased HSCs in the niche [30]. MSC populations in the periarteriolar niche, including NG2+ MSCs, have different effects on maintaining HSC function. However, Asada et al. demonstrated that the main source of SCF to maintain HSCs in the BM was sinus gland LEPR+ CAR cells, not the NG2+ cells in the periarteriolar niche [27]. Similarly, experiments in LEPR-Cre mice confirmed the importance of SCF in MSCs [32]. MSCs were found to secrete CXCL12 and many other factors (e.g., SCF, VCAM-1, angiopoietin-1, IL-6, IL-11, thrombopoietin (TPO), fms related receptor tyrosine kinase 3 (Flt-3) ligands, granulocyte colony stimulating factor (G-CSF), granulocyte–macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), and BMP4), which typically affect HSC populations and their hematopoietic function [32, 33]. In addition, deleting CXCL12 negatively affected the HSC population by redistributing the remaining HSCs population in the BM, whereas depleting CXCL12 in the LEPR+ MSC of the sinus gland did not affect the HSC population. Nes+ MSCs are located extremely close to Schwann cells and participate in the mobilization of HSCs via the sympathetic nervous system (SNS) [34]. In contrast, the knockout of nestin decreased the number of MSCs and increased HSC proportions within the spleen [35]. Furthermore, the co-cultivation of MSCs with HSCs increases the survival and expansion of HSCs within the spleen [35]. Additionally, the role of periarteriolar stromal cells in resting-state HSCs was confirmed by the recovery of mice with myelosuppression, polyinosinic-polycytidylic acid therapy, or Pml knockout, all of which significantly altered the distance between HSCs and arterioles, allowing HSC overproliferation, HSC population depletion, and migration from the arterioles [34, 36]. Many studies have demonstrated the role of osteoblast lineage cells in HSCs, indicating that hematopoiesis regulation by osteoblasts depends on their differentiation stage [37]. Self-renewing osteoprogenitors in the BM can also form a supportive HSC niche [38]. Nes+ MSCs have been demonstrated to produce CXCL12 and SCF and colocalize with HSCs, and even lead to HSC mobilization when the numbers of Nes+ MSCs are reduced, suggesting that osteogenesis and HSC maintenance in the BM are negatively associated [6].

Studies have also demonstrated the involvement of the early B cell factor (Ebf) in B-cell differentiation and bone development [39], as well as the preferential expression of Ebf3 (an Ebf family member) in CAR cells [40]. Using a conditional knockout mouse model, the specific knockout of Ebf3 expression in CAR/LEPR+ cells was found to significantly reduce the number of long-term repopulating HSCs, pre-erythroid cells of erythroid progenitors (MEPs), common lymphoid progenitors, and granulocyte/monocyte progenitors, as well as reduce the number of functional HSCs and cause the increased differentiation of most Ebf3/ CAR cells into the osteoblastic lineage [40]. A researcher has identified CAR cells subtypes (i.e., Adipo-CAR and Osteo-CAR cells) using single-cell sequencing and determined the localization of these subtypes and elucidated the source of pro-hematopoietic factors [29]. What’s more, a study has revealed that the proportion of CAR cells in ClC-3/ mice increased by 14.28%, whereas the number of HSCs decreased by 11.81% [41]. Additionally, the expression of the transcription factor Ebf3 was decreased, while the ECM receptor signaling pathway was enhanced, suggesting that ClC-3 chloride channels may regulate Ebf3 expression, thereby mediating the participation of the HSC niche in the bone remodeling process [40, 41]. Overall, the osteogenic and adipogenic differentiation potentials of CAR cells are inhibited by the expression of the transcription factors Ebf1/3, ensuring that these specialized MSCs remain undifferentiated and allowing for marrow cavity maintenance by controlling CXCL12 and SCF expression as well physically preserving the HSC niche [28].

Osteoblasts and hematopoietic stem cells

Osteoblasts are important components of the HSC niche [28]. These cells localize as anchored cell populations within the bone, producing large amounts of extracellular proteins, such as type I collagen, osteocalcin (OCN), and alkaline phosphatase [29]. A recent in vitro co-culture study showed that osteoblast adhesion favored HSC self-renewal [42]. Moreover, CFU assays and thymidine suicide techniques demonstrated the enrichment of HSPCs near the intraosseous niche [3]. Osteoblasts also synthesize and express many HSC-influencing factors, such as TPO, angiopoietin-1, and osteopontin, which prevent the increase in the HSC population [28, 43]. As mentioned earlier, SDF-1/CXCL12 and SCF, which are extrinsic factors released by HSC-supporting cells, are important regulators of HSC maintenance and retention in the BM [5, 9].

Additionally, NCAD+ osteoblasts on the endosteal surface of the bone can interact with HSC precursors via a mechanism that may support HSCs through the BMP signaling pathway [24]. In contrast, the ablation of NCAD+ osteoblasts has been shown to cause irreversible changes in HSC function during homeostasis and regeneration [22]. NCAD+ osteoblasts can also control HSC status and function through noncanonical Wnt signaling [22, 44]. Furthermore, osteoblast ablation results in decreased HSC proportions in the bone with a concomitant increase in extramedullary hematopoiesis, whereas an increase in osteoblast numbers leads to increased HSCs [15, 28]. Interestingly, osteoblasts not only affect HSCs but also modify their well-differentiated progeny [28]. Osteoblasts influence the status of silenced HSCs through the Tie2+/angiopoietin-1 and TPO/MPL signaling pathways, which facilitate the interaction between HSCs and niche components that promote quiescence and self-renewal of these cells [33, 45].

These results show that although the proportion of osteoblasts is important for maintaining HSC function, it is not the only determining factor [42]. Polycomb genes can regulate cell fate, including Bmi-1 and Mel-18 [42]. When HSCs divide, self-renewal cells attached to osteoblasts have high Bmi-1 expression and low Mel-18 expression and when stem cells leave the osteoblastic niche undergo differentiation, while the expression of the aforementioned proteins is opposite in differentiated cells [42]. Osteoblasts also play an important role in regulating HSC stress response. For example, injecting mature osteoblasts with 5-fluorouracil (5-FU) led to cell death, along with reduced osteoblast gene expression in the stromal cells and increased wnt family member 5a (Wnt5a) expression in some osteoblasts [46]. This increased expression of Wnt5a in these bone lineage cells may reduce the proliferation rate of HSC subsets, thereby protecting them from DNA damage and apoptosis after 5-FU chemotherapy [47]. The specific mechanism involves the interaction of Wnt5a with Ryk, a Wnt ligand receptor, to maintain HSC quiescence and protect HSCs after myeloablative stress [47]. However, 5-FU did not exert any influence in the intraosseous niche of aged mice [48], suggesting that stress response changes with age. Overall, the synthesis and expression of osteoblasts are influenced by HSCs, while osteoblasts mediate the self-renewal and differentiation direction of HSCs through extracellular proteins, cytokines, signaling pathways, stress responses, and proportions.

Endothelial cells and hematopoietic stem cells

Endothelial cells are a major type of BM non-hematopoietic cells that create an HSC niche primarily involved in controlling hematopoietic cell trafficking and homing [49]. Early study has found that human BM endothelial cells support the proliferation of hematopoietic cells in vitro cultures, indicating the role of endothelial cells in maintaining hematopoietic progenitor cells [50]. Furthermore, these cells are located in the niche and in direct contact with HSCs [11]. Moreover, the co-transplantation of endothelial progenitor cells with HSCs has been revealed to promote endothelial cell recovery as well as hematological and immunological reconstitution [11]. Studies investigating endothelial cells in the HSC niche have mainly focused on arteriolar endothelial cells (AECs) and sinusoidal endothelial cells (SECs) [51].

These two groups of cells function differently, but both cell types produce SCF required for hematopoietic stem cell survival [51]. AECs, along with CAR cells synthesize the ECM protein Del-1, block SEC regeneration by producing neutralizing antibodies against Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) or VE-cadherin and prevent hematopoietic cell reconstitution [52, 53]. Conversely, SECs synthesize only a small fraction of Del-1 [54]. Moreover, SECs are required for regenerative hematopoiesis, dependent on VEGFR2 for their regeneration, and produce high amounts of CXCL12 and E-selectin [54]. Additionally, the deletion of CXCL12 or SCF in Tie2+ endothelial cells has been shown to reduce HSC numbers [11, 49, 55]. Early study has found that human BM endothelial cells support the proliferation of hematopoietic cells in vitro cultures, indicating the role of endothelial cells in maintaining hematopoietic progenitor cells [50]. Furthermore, Kiel et al. demonstrated an association between HSCs expressing CD150, CD48, and CD41 and SECs [56].

HSC proliferation is promoted via the expression of E-selectin (exclusively found only on endothelial cells), whereas E-selectin antagonists induce HSC dormancy and self-renewal [54, 57]. Studies have suggested that the endosteal niche provides a hypoxic environment that maintains HSCs in a dormant state, whereas the vascular niche enables the proliferation and differentiation of HSCs in aits comparatively higher oxygen-containing environment [58, 59]. Lampreia et al. demonstrated that endothelial cells also stimulate HSC self-renewal by synthesizing Notch ligands [60]. In particular, the conditional deletion of Jagged-1, an endothelial cell-generated Notch ligand, in the endothelial cells causes depletion of the HSC pool and a severe decline in hematopoiesis [60, 61]. Jagged-1 is also involved in the regeneration of hematopoiesis after myeloablation [61, 62]. Lastly, erythroid precursors have been reported to differentiate into endothelial cells to restore the endothelial cells necessary for vascular network production [63]. Besides, endothelial cells and LEPR cells produce high levels of soluble SCF in the early postnatal bone marrow [55]. Membrane bound SCF from endothelial cells promotes the maintenance of HSCs in the early postnatal bone marrow, while SCF from LEPR cells is used to maintain HSCs and restricted hematopoietic progenitor cells in adult bone marrow [55]. Thus, co-culturing endothelial cells with hematopoietic progenitors may contribute to the expansion of the latter cells, helping to maintain HSCs in the niche.

Megakaryocytes and hematopoietic stem cells

MKs are platelet-producing cells mainly found in the BM [64]. These cells are important regulators of hemostasis and vascular integrity, as well as key components in thrombosis and inflammatory responses [65].However, MK function is not restricted to platelet production. Complex experimental studies in recent years have suggested that MKs are crucial regulators of BM HSCs. Bruns et al. demonstrated that MKs govern quiescence in the HSC niche [66]. Gene expression analysis studies have revealed that MKs in the BM are a source of chemokine motif ligand 4 (CXCL4, also known as platelet factor 4 [PF4]), which regulates HSC cell cycle activity [66]. In line with this finding, injecting mice with CXCL4 decreased HSC numbers due to their increased quiescence, whereas CXCL4−/− mice exhibited increased HSC numbers and proliferation [67].

Another study also demonstrated the association between HSCs and MKs, showing that MK ablation leads to HSC proliferation [68]. The ablation of MKs results in decreased levels of the biologically active transforming growth factor (TGF)-β1 protein in the BM and reduced levels of nuclear-localized phosphorylated SMAD2/3 (pSMAD2/3) in HSCs [69]. These findings suggest that MKs mediate HSC cell cycle activity via TGF-β1, and the absence of TGF-β1 conditions in MKs increases HSC activation and proliferation [68,69,70]. Thus, TGF-β1 as a major signal generated by MKs to maintain HSC quiescence. Consequently, considering that the MK ablation or loss of TGF-β1 conditions in MKs induce quiescence in HSCs, MKs involved in the maintenance of HSC quiescence. Similarly, further studies confirmed the loss of HSC quiescence after MK depletion and indicated that TPO secreted by MKs was involved in HSC maintenance [71]. In addition to the secretion of CXCL4 and TGF-β1, MKs are major producers of TPO [66, 70, 71]. Moreover, extensive studies have shown that TPO and its receptor c-MPL regulate hematopoietic activity [71, 72]. A study on MK-depleted mice found that reduced MK production led to a reduction in both the number and quiescent phase of HSCs [64]. The disturbance of HSC quiescence in the BM was achieved by ablating the MK populations in the mice [64]. Furthermore, destroying MKs may decrease TPO concentrations within the BM because of the production of TPO by MKs [72]. This concept was supported by the regaining of HSC function in TPO-injected MK-ablated mice.

However, considering MKs may control HSC proliferation through secreted proteins, the calcium ion-dependent C-type lectin-like receptor 2 (CLEC-2) may stimulate MKs to produce TPO [73]. Therefore, direct contact between MKs and HSCs may not be essential. Additionally, Ludin et al. have shown that a subset of α-smooth muscle actin (SMA)+ monocytes and macrophages maintain HSPCs [74]. It is important to determine the interaction of HSCs with megakaryocytes or α-SMA+ monocytes and macrophages by deep confocal imaging of α-catulin-GFP+-c-kit+ HSCs [74, 75]. So, MK cells can indirectly mediate HSC proliferation through the secretion of extracellular proteins CLEC-2 and SMA. Under stress conditions, fibroblast growth factor 1 (FGF1) signaling in MKs temporarily dominates TGF-β inhibitory signaling to stimulate HSC proliferation [76]. Furthermore, MKs close to HSCs are activated by CXCL4, whereas TGF-β1 secretion regulates their quiescence [67]. Thus, FGF1 produced by MKs also promotes HSC expansion in stress conditions. All these observational results indicate that MK is an ecological niche cell derived from HSC, which can directly or indirectly dynamically regulate the function of HSC.

Macrophages and hematopoietic stem cells

Similar to other niche components, macrophages directly influence HSCs in the BM niche, and macrophages in the splenic niche retain HSCs via VCAM-1 adhesion molecules [77]. Macrophages also have a unique and indispensable role in the organization of BM erythroid islands, a highly specific erythropoietic niche that contains a specialized central macrophage surrounded by erythrocytes [78]. In these structures, the central macrophage promotes erythroblast maturation in several ways, including mitochondrial clearance by tunneling nanotubes [79]. Several studies have established that G-CSF treatment leads to HSC mobilization, production of granulocytes, inhibition of macrophages and osteoblasts [80, 81], and activation of norepinephrine secretion by sympathetic neurons of the BM microenvironment [82].

Certain macrophage populations in the BM have been found to exhibit α-SMA and increased expression of the cyclooxygenase (COX)−2 [83]. These macrophages can resist radiation-induced cell death and further upregulate COX-2 expression in response to stress [84]. In this mechanism, COX-2-derived prostaglandin E2 (PGE2) may limit reactive oxygen species production by inhibiting the Akt kinase and activating the increased expression of the stromal cell chemokine CXCL12, thereby preventing HSPC exhaustion required for HSC quiescence [74].

Another function of macrophages is to inhibit HSCs from entering the quiescence phase via the interaction of CD82 with HSCs through the Duffy antigen receptor for chemokines (DARC)/CD234 on the macrophages [85]. This interaction activates TGF-β1/SMAD signaling and inhibits the proliferation and differentiation of the HSCs [85]. Moreover, macrophages have been recognized as a pool of cells with regulatory properties because they exert humoral effects on Nes+ cells via unidentified cytokines and induce these cells to secrete CXCL12, thereby maintaining HSCs in the niche [84]. Furthermore, macrophages have been suggested to promote HSC retention in their BM niches via Nes+ stromal cells, as evidenced by the macrophage depletion in the BM causes HSC mobilization in the blood [80]. From this, it can be seen that macrophages directly affect the HSC niche, retaining HSCs through VCAM-1 adhesion molecules and maintaining their quiescence through stress response.

Lymphocytes and hematopoietic stem cells

A subset of early lymphoid progenitors has been reported to reside in the endosteal niche and is maintained by factors produced by osteoblasts [86]. Other lymphoid progenitors are found in the perivascular niches created partly by LEPR+ cells [9]. Among the numerous lymphocytes colocalizing with HSCs, Foxp3+ regulatory T (Treg) cells play a significant role in protecting HSCs from immune attack in the niche [9]. In particular, allogeneic HSCs (as well as syngeneic HSCs) survive in unirradiated mice for at least 1 month after transplantation, whereas the depletion of Treg cells results in the rapid loss of allogeneic HSCs [87]. Furthermore, Treg cells in the BM show elevated production of IL-10, which directly affects MSC function [88, 89]. Thus, IL-10 secreted by Treg cells regulates hematopoiesis via another mechanism, indicating that this cytokine is required for HSCs maintenance [89]. Moreover, BM Treg cells are migratory in nature and home in the BM [39]. After Treg cell depletion, the function and phenotype of MSCs and HSCs are impaired. This Treg depletion-associated impairment of MSC and HSC functions results in the reduced hematopoietic supportive capacity of the niche [89]. HSPCs are also lost after Foxp3+ Treg cell depletion [87]. This loss of HSPCs may be linked with the clear colocalization of HSPCs with Treg cells and their accumulation at the endosteal surface of the calvaria and trabecular bone.

Moreover, the highly CD150-expressing subpopulation of Treg cells in the BM produced elevated levels of extracellular adenosine via the cell-surface ectoenzyme CD39 [90]. The generated adenosine then enhances Treg cells activity, protects HSCs from oxidative stress, and maintains HSC quiescence [39]. Furthermore, the co-transplantation of this Treg cell subset, instead of other Treg cell subsets, was found to promote better engraftment of HSCs in allogeneic hosts [90]. Studies have indicated that perisinusoidal LEPR+ cells seem poised to undergo adipogenic differentiation, whereas periarteriolar LEPR+ cells appear poised to undergo osteogenic differentiation [29, 46, 91]. The adenosine produced by these LEPR+ cells upon differentiation may act in opposite directions by activating immunity and decreasing the immune privilege of the niche [92]. However, scarce evidence exists on the potential role of B lymphocytes in the BM niche. Nevertheless, recent studies have suggested that acetylcholine, a neurotransmitter abundantly produced by B cells, limits hematopoiesis in vivo [93].

Therefore, Treg cells and B lymphocytes in the BM niche can be considered as immune privileged sites and protective zones for HSCs. Furthermore, Treg cells can create a privileged zone in the bone marrow, protecting HSCs from oxidative stress and keeping them quiescent to protect HSCs in the niche after transplantation.

Adipocytes and hematopoietic stem cells

Adipocytes are the most abundant stromal components of the adult BM and are considered negative hematopoiesis regulators [94]. There is a well-established interrelationship between hematopoiesis and adipose tissue within the human BM [95]. This understanding is derived from the fact that the red BM at birth is largely devoid of adipocytes and exhibits highly active hematopoiesis [95]. In contrast, the red marrow, when replaced with yellow marrow in adults, is enriched in adipose tissue and characterized by reduced hematopoietic activity [96]. After bone marrow transplantation (BMT), the destruction of hematopoietic function in HSCs by mouse bone marrow adipocytes via affecting lipid raft/TGF‐β signaling pathway [95]. Furthermore, this view was supported by findings showing that hematopoietic recovery after irradiation is significantly accelerated in mice unable to produce adipocytes or after adipogenesis inhibition by PPAR-γ antagonists [97].Additionally, Wilson et al. have demonstrated that PPAR-γ knockout mice devoid of adipocytes display severe extramedullary hematopoiesis [98]. This finding may be related to the observed dysregulation of the CXCL12/CXCR4 axis, implying that adipocytes may be involved in HSC retention or mobilization [98]. One such related mechanism may be a positive feedback loop in which adipocytes, via the strong secretion of monocyte chemotactic protein-1 (MCP-1), stimulate MSC differentiation into new adipocytes and negatively affect HSCs [99].

Numerous studies have revealed that niche components produce adiponectin, while HSCs express adiponectin receptors [100]. Moreover, adiponectin increases HSC proliferation via a p38 MAPK-dependent pathway and maintains their undifferentiated state [101]. Consequently, adiponectin deficiency in mice impairs the ability to restore hematopoiesis after chemotherapy [101]. Moreover, after MSC exposure to radiotherapy or chemotherapy, the adipocyte numbers increase due to enhanced adipogenic differentiation of the MSCs [102]. This substantial increase in the number of adipocytes hinders hematopoietic regeneration, which can be used as diagnostic criteria for BM hypoplasia [102]. Although leptin, another adipokine secreted by adipocytes independently, exerts a negligible effect on the survival and proliferation of mouse and human HSCs in vitro [103]. Leptin exhibits its pleiotropic effects on MSC differentiation into adipocytes in vivo [103]. In addition, leptin demonstrates a synergistic effect with SCF by stimulating the proliferation of primitive hematopoietic progenitor cells in vitro during the formation of HSCs and progenitor cell colonies [55].

In the BM, adipocytes produce hematopoietic factors, including CXCL12, IL-8, colony-stimulating factor 3 (CSF3), and leukemia inhibitory factor (LIF), comparable to MSCs [104]. Furthermore, BM adipocytes have been shown to promote HSC regeneration and hematopoiesis after irradiation or 5-FU treatment [105]. Adipocytes and their precursors, which comprise a small LEPR+ cell subpopulation, produce the required SCF for hematopoietic recovery [105]. SCF from these LEPR+ cells (but not from endothelial cells or osteoblasts) also activate regeneration, whereas the conditional deletion of SCF in the adipocytes inhibits hematopoietic regeneration [105]. In contrast, in previous comparative studies, obese mice showed a significant increase in lymphocytes and adipocytes in the bone marrow, leading to an increase in the expression of leptin mRNA in bone tissue, indicating that bone marrow adipocytes enhance hematopoietic function by secreting leptin [98]. Similarly, high-fat diet induced obesity leads to an increase in the number of bone marrow precursor cells in HSCs [96]. Thus, adipocytes formed under hematopoietic stress create large amounts of leptin and SCF, representing an emergency response that provides the factors necessary for the survival and expansion of HSCs.

Neural and hematopoietic stem cells

Autonomic nerve fibres extensively innervate the bone and BM. The physiological role of the SNS involves regulating bone formation, hematopoiesis, and cancer progression [106, 107]. Furthermore, the BM is co-innervated by the SNS and sensory nerves, which envelop the arterioles and directly contact periarterial NesGFP+ stromal cells [108]. Méndez Ferrer et al. further showed that SNS neural connectivity plays a key role in the BM, including the mobilization of HSCs from the BM via circadian rhythms [109]. In this mechanism, sympathetic nerve fibres release the neurotransmitter norepinephrine, which exerts its influence via the β3 adrenergic receptors [110]. The activation of the SNS exerts control on HSCs from the BM and downregulates CXCL12 levels during the daytime, leading to activation of HSC activation and enhanced production of neutrophil and inflammatory monocytes [111]. The SNS may also play a multidimensional role in the homeostatic BM by targeting different cellular components [111]. The bone is innervated by fibres of the parasympathetic nervous system (PNS) and considering that the PNS and SNS are known to have opposing roles in most organ systems, the PNS may antagonize sympathetic regulation in the bone (and possibly the BM) [112]. The parasympathetic cells produce the neurotransmitter acetylcholine that transmits signals via nicotinic or muscarinic receptors [113]. Researchers have demonstrated that the parasympathetic signals promote bone mass accumulation by mediating nicotinic receptor activation, regulating osteoblast proliferation, and osteoclast apoptosis [114].

Moreover, the cholinergic nervous system counteracts the sympathetic noradrenergic system by inhibiting it at night, thereby reducing HSC outflow [115]. Additionally, the cholinergic system maintains HSC quiescence in the endosteal niche during proliferative stress via CXCL12 production [116]. However, the type 1 muscarinic receptor (CHRM1), a receptor for the parasympathetic neurotransmitter acetylcholine in the hypothalamus, has been found to regulate G-CSF-induced HSC mobilization from the BM through glucocorticoid release [117].

Glial cells, which include Schwann cells, in the PNS envelop the axons of the peripheral nerves [118]. Schwann cells consist of myelinating and non-myelinating cells [119]. The myelinating Schwann cells that supply myelin to the nerves express myelin basic protein (MBP) but not glial fibrillary acidic protein (GFAP) [120]. In contrast, non-myelinating Schwann cells (which wrap around the sympathetic nerves in the BM) express GFAP but not MBP [120]. Schwann cells are part of the BM niche, producing functionally active TGF-β and leading to the phosphorylation of the SMAD2/3 signaling pathways that affect cell cycle progression [121]. During this process, regulating the activation of latent TGF-β signaling promotes HSC quiescence and maintenance directly in the BM [121].

Furthermore, Wallerian degeneration and the clearance of GFAP+ Schwann cells in the BM were shown to be induced by the transection of postganglionic sympathetic nerves [122]. Additionally, decreased neuregulin-1 signaling due to nerve injury can trigger Schwann cell apoptosis [123]. Moreover, non-myelinating Schwann cells were demonstrated to have the most prominent role in the HSC quiescent phase, with the loss of the autonomic nerves of the BM reducing the number of these secreting Schwann cells and leading to the disappearance of HSCs [124]. Therefore, the excitation of the sympathetic nervous system controls the rhythm of HSC mobilization, while the cholinergic nervous system promotes the stillness and maintenance of HSCs in BM by regulating TGF-β signaling.

Crosstalk between osteoblasts and osteoclasts

Extracellular vesicles in osteoblasts (also known as small osteoblast vesicles [SOVs]) are secreted and trapped by mature osteoblasts (MOBs) [125]. Studies have indicated that a fraction of MOB-derived SOVs limits bone formation and stimulates osteoclast genesis [125]. Moreover, the intercellular communication between osteoblasts and osteoclasts is enabled by SOVs via the upregulation of RANKL in SOV-treated osteoblasts [126]. These results explain the negative regulation of bone formation by SOVs through MOBs and the positive regulation of bone resorption, thus facilitating the ‘‘reciprocal reversal phase’’ via microRNA-mediated mechanisms [126]. Furthermore, SOVs not only have a direct effect on osteoclasts but also exhibit a direct effect on the BM [127]. For example, the inhibition of osteoblast production and enhanced osteoclast genesis in the BM have been revealed in HSCs co-cultured with SOVs [127]. Moreover, osteoclasts were demonstrated not to affect HSC maintenance in osteoclast differentiation models with cytokine deficiency as well as in c-Fos-deficient and RANKL-deficient mice models [126, 128, 129]. However, osteoclasts were found to promote HSC migration from the BM into the blood circulation when the endosteum was disrupted [129].

Osteoclast–osteoblast communication occurs in basic multicellular units (BMUs) during the initiation, transition, and termination phases of bone remodeling. During the initiation phase, hematopoietic precursors are recruited to BMUs [130, 131]. These precursors then express cell surface receptors, including c-Fms, RANK, and osteoclast-associated receptors, followed by their differentiation into osteoclasts after contact with ligand-expressing osteoblasts [132, 133]. Osteoclasts and osteoblasts communicate using three pathways: cell–cell contact, diffusible paracrine factors, and the cell-bone matrix [134]. Furthermore, EphrinB2 expression on osteoclasts and EphB4-mediated bidirectional signaling in osteoblast precursors are important components of osteoclast–osteoblast communication [135]. Reverse signaling in osteoclasts through EphrinB2 decreases c-Fos and NFATc1 activity and inhibits osteoclast function, whereas positive signaling in osteoblast precursors via EphB4 reduces RhoA activity, thereby enhancing osteoblast differentiation [136]. These interactions most likely occur during the termination phase in the bone remodeling compartment, where bone remodeling is achieved by osteoblastic bone formation and bone matrix mineralization (Fig. 3).

Fig. 3
figure 3

Schematic illustration of the evolution of osteoblasts and osteoclasts during bone formation and bone remodeling. HSCs first evolve into mononuclear, then into fused polykaryon, and finally form osteoclasts through the RANKL pathway and OPG mediation, achieving bone resorption. MSCs first evolve into preosteoblasts, then osteoblast progenitor cells, and finally form osteoblasts, achieving bone formation

Crosstalk between MKs and bone cells

The lack of MKs can substantially impair bone formation. Further research suggests that the increased secretion of TGF-β by MKs promotes the proliferation and differentiation of osteoblasts, as well as the formation of CD31+ Emcn(Endomucin)+ blood vessels [69]. Additionally, treatment with MKs or TPO significantly mitigates radioactive bone injury in mice via the direct or indirect increase in TGF-β1 levels in the BM [70]. MK-derived TGF-β1 is also involved in inhibiting apoptosis and promoting DNA damage repair in radiation-exposed osteoblasts [70].

However, the effect of MKs on osteoblast differentiation remains controversial because MKs have been shown to inhibit osteoblast differentiation. Recently, Lee et al. demonstrated that an MK-conditioned medium could promote osteoblast amplification while inhibiting its differentiation [137], partly by modulating Pyk2 phosphorylation, a negative regulatory process of osteoblast differentiation [138]. Furthermore, MKs migrate to their ecological niche in the bone through TPO signals and secrete platelet-derived growth factor receptor-β (PDGF-β) to promote osteoblast expansion and ecological niche remodeling [139]. In addition, MKs inhibit osteoblast differentiation to maintain its hematopoiesis-enhancing activity (HEA) during steady-state conditions [140].

Studies have shown that MKs inhibit bone resorption and exert bone-protective effects by stimulating bone formation, making MKs an ideal therapeutic target for metabolic bone diseases. Moreover, immature osteoblasts have higher HEA than mature osteoblasts [37, 141]. In vitro studies in aged mice have demonstrated that MKs inhibit the differentiation of osteoclast precursors into osteoclasts, thereby curbing bone resorption [137, 142]. Furthermore, the mice exhibited a decrease in the MK potential to stimulate osteoblast expansion despite the increase in the number of MKs [143]. Facts prove that hematopoietic ecological niche remodeling was necessary for increased success in HSC transplantation after total-body irradiation in mice [144].

Osteomacs (OMs) have been reported to play a role in increased mouse survival after irradiation, with an interaction between OMs and MKs in the irradiated HSC transplantation [69]. Interestingly, MKs require physical contact with OMs rather than HSPC support to enhance their effectiveness [69]. In support of this finding, some studies have suggested that MKs regulate the hematopoietic niche through protein secretion and physical contact [145]. Furthermore, the number of OMs and osteoblasts in aged mice also decreased, along with a decrease in HSC transplantation and myeloid bias [146]. Finally, MKs will likely lose their HEA potential even though the number of MKs increases.

Crosstalk between lymphocytes and bone cells

Lymphocytes mediate the regulation of osteoblast maturation and activation. IL-17A and IL-17F secreted by Treg cells strongly promote osteoblast differentiation in MSCs after binding to bone morphogenetic protein-2 (BMP-2) [17, 147]. Researchers have revealed that some cytokines, including interferon-γ secreted by CD4+ T lymphocytes, can promote the differentiation of MSCs into osteoblasts [148]. Furthermore, the activity of TGF-β, which is also produced by CD4+ T lymphocytes, negatively correlates with osteoblast differentiation [149]. Both patients with osteoporosis and oophorectomy (OVX)-induced osteoporosis animal models demonstrate increased TNF-α expression in T cells and osteoblast apoptosis [150]. Additionally, osteoblasts may play a role in antigen presentation, activating T cells as helper cells and promoting T cell differentiation and maturation [150].

Similarly, B cells also exhibit a regulatory effect on osteoblast function. Studies on rheumatoid arthritis have found that B cells produce osteoblast-inhibiting factors, such as CCL3 and TNF, and activate the ERK and NF-κB signaling pathway that inhibits osteoblast differentiation, thereby suppressing bone formation in RA [151, 152]. In addition, RANKL produced by B cells promotes osteoclast production, directly disrupts bone metabolism balance, and causes postmenopausal osteoporosis [152, 153]. In an in vitro model of human osteoclastogenesis, human B lymphocytes have been shown to stimulate osteoclastogenesis inhibitory factor (OPG) production by secreting TGF—β, thereby inhibiting osteoclast formation [152]. Compared with young mice, B lymphocytes isolated from bone marrow and spleen produce significantly higher levels of RANKL and OPG in elderly mice [152]. In addition, Notch signaling transduction is also considered a mediator of the impact of activated B lymphocytes on osteoblast generation [153]. Conversely, IL-7 and CXCL12 secretion by osteoblasts can induce B-cell differentiation in HSCs [154].

In addition, lymphocytes regulate the chondrocyte differentiation process by secreting certain cytokines. Chondrocytes mainly produce large amounts of ECM to form cartilage and preserve normal cartilage function [155]. Additionally, IL-6 secreted by activated T cells can induce the production of tissue inhibitors of metalloproteinases, thereby playing a role in cartilage protection [153]. Furthermore, Kuhl et al. demonstrated that IL-3 secreted by T cells could increase the expression of the chondrocyte-specific genes sex-determining region of Y chromosome (SRY)-box transcription factor 9 (SOX9) and type IIa collagen [155]. SOX9 is a key transcription factor in chondrocyte development and maturation, participating in the entire differentiation process of chondrocytes [156]. Researchers have also found that TGF-β positively influences cartilage remodeling [157]. Studies have shown that T cell-secreted TGF-β can increase the mRNA expression of chondrocyte differentiation-related genes (including SOX9) and facilitate chondrocyte differentiation [158]. In contrast, chondrocytes can directly inhibit T lymphocyte proliferation, which restricts the expression of the IL-2 receptor α-chain (CD25) in CD4+ T cells [159].

Crosstalk between macrophages and mesenchymal stem cells

In the BM niche, macrophages are key regulators in inflammatory response initiation, propagation, and resolution phases during tissue regeneration [34]. Macrophages promote HSC retention by regulating MSCs and, along with MSCs, can co-regulate the physiological processes in the body [160]. Macrophages have been reported to have an extensive adaptive phenotypic and functional shift that may exacerbate and resolve inflammation during tissue repair [161, 162]. The mechanism of macrophages in bone healing has been gradually elucidated over recent years, with results indicating the presence of three main types of niche macrophages: M0, M1, and M2 type [163, 164]. M0 macrophages exhibit a strong ability to enhance the osteogenesis of MSCs [163]. M1 macrophages only participate in the early osteogenic phase, an inflammatory response regulated by MSCs in the early phase of bone injury, and are absent in the later phase of bone mineralization [163]. Compared with M1 macrophages, M2 macrophages release more BMP-2, stimulating the nuclear transfer of RUNX2 by activating the SMAD1 signaling pathway and upregulating ALP and OCN expression in MSCs, consequently inducing MSCs to form thick cell sheets [164]. Moreover, MSCs support osteogenesis promotion by M2 macrophages, which in turn is conducive to the osteogenic differentiation of MSCs [165]. Therefore, M2 macrophages have a relatively greater ability to promote tissue regeneration.

MSCs are the main functional cells of the bone healing process [166]. The upregulation of CD54 production in MSCs after contact with M1 macrophages increases the inhibitory effect on T-cell proliferation [167]. Moreover, MSCs can induce immune tolerance through TSG-6-dependent paracrine effects and the intercellular interactions between MSCs and proinflammatory macrophages [168]. Macrophage-derived oncostatin M has been reported to mediate crosstalk between NesGFP+ MSCs and macrophages [169]. In general, M1 macrophages trigger apoptosis of MSCs and inhibit MSC growth in vitro, whereas M2 macrophages support MSC growth, proliferation, and migration [170].

New immunotherapies for bone tumors in the BM niche

Osteosarcoma

Osteosarcoma (OS) is a common primary malignant bone tumor that usually occurs at the metaphyseal end of a long bone and is clinically characterized by localized pain and swelling [171]. The age of onset of osteosarcoma has a bimodal pattern, with incidence peaking in the second decade of life and late adulthood, with a 5-year survival rate of approximately 70% without metastases and 30% at the time of diagnosis [172]. The current standard of care for osteosarcoma includes chemotherapy, amputation, etc., and although chemotherapy has significantly advanced the treatment of osteosarcoma, the 5-year survival rate in drug-resistant forms of osteosarcoma is still less than 20% [173]. In recent years, the rise of immunotherapy has provided a new strategy for the treatment of osteosarcoma [173].

The tumor microenvironment (TME) of bone tumors interacts with the BM niche [171]. In OS, osteoclasts control the occurrence of osteoclasts and coordinate bone remodeling through the RANKL/RANK/OPG signaling pathway [173]. Recent studies have shown that cGAS/STING signaling can promote the production of type 1 interferon and enhance the antigen presentation of T cells, which can exert immunostimulatory effects [174]. Cytokines such as IL-2 activate natural killer cells (NK) and cytotoxic T lymphocytes (CTLs) to enhance the immune system's attack on tumors and are used to treat osteosarcoma [175]. About cytotoxic T cell lymphocyte antigen 4 (CTLA-4) expressed on regulatory T cells (Tregs), is a co-inhibitory cytokine of the B7 ligand of the CD28 receptor during T cell activation [176]. In addition, the anti-PD-1 antibody durvalumab (ClinicalTrials.gov ldentifiers: NCT02815995) and the anti-PD-L1 antibody nivolumab (ClinicalTrials.gov ldentifiers: NCT02304458) are used to restore the anti-tumor activity of T cells by blocking the PD-1/PD-L1 pathway, thereby achieving the goal of treating osteosarcoma [177]. Tumor-associated macrophages (TAMs) are the main tumor-infiltrating immune cells of TME. Macrophages, which are part of BM niche, are associated with poor prognosis when their expression genes are elevated, resulting in increased infiltration of TAMs [178]. Current targeted therapies for TAM such as inhibition of signaling pathways such as CSF-1/CSF-1R, CCL2/CCR2, and CD47/SIRPα are still being evaluated clinically, and further studies are needed to determine their translational benefit in sarcoma patients [179].

In recent years, a research group has confirmed the tumorigenic effect of ephrin type-A receptor 2 (EphA2) in OS cells, with higher expression in tumors with higher Huevo’s grade than lower ones, and higher expression in OS cells with worse prognosis and higher recurrence rate [180, 181]. The activity of EphA2 was inhibited by treatment with two receptor tyrosine kinase inhibitors, pazopanib and trametinib [180, 181]. This finding revealed that the downregulation of EphA2 expression had a significant inhibitory effect on cell proliferation and migration [181]. In addition, studies have shown that HER2 is a protein on the surface of OS cells, and that T cells have been genetically engineered to become CAR cells that express HER2 and that these CAR-T cells are able to specifically recognize and kill HER2-expressing OS cells [181, 182]. An antibody–drug conjugate consisting of trastuzumab, a monoclonal antibody against HER2, and deruxtecan, a topoisomerase I inhibitor, is being evaluated for the treatment of recurrent osteosarcoma [182]. These advances have shown the potential of immunotherapy in the treatment of osteosarcoma, and although the current efficacy of immunotherapy in osteosarcoma is still limited, these studies provide the basis for possible therapeutic breakthroughs in the future.

Chondrosarcoma

Chondrosarcoma (ChS) is a malignant tumour that originates in chondrocytes, and it is the second most common type of osteosarcoma[183]. Although surgical resection is the preferred treatment for ChS, conventional chemotherapy and radiation therapy often do not respond well to tumors that have a high metastatic potential or are difficult to remove by surgery, resulting in a generally poor prognosis [184]. As a result, the rise of immunotherapy has provided an emerging therapeutic target for ChS.

Recent studies have found that the expression level of PD-L1 in ChS may be closely related to the aggressiveness of tumors and the survival rate of patients [185]. This finding suggests that PD-1/PD-L1 inhibitors are promising as a new strategy for the treatment of ChS. ChS cells are able to use certain mechanisms to evade recognition and attack by the immune system, which may lead to alterations in the tumor microenvironment that would otherwise be immunogenic [177, 185]. TAMs in ChS cells induce immunosuppression by expressing the colony-stimulating factor 1 receptor (CSF1R), also known as CD115 [184]. Activation of CSF1R is mediated by two ligands secreted by ChS cells: CSF-1 and IL-34 [179]. The involvement of these ligands leads to the release by macrophages of tumor growth-promoting cytokines, such as TGF-β and IL-10, which further transform the immune microenvironment into a state conducive to tumor growth, making it more susceptible to progression and recurrence [179]. Similarly, the expression of EphA2 was significantly higher in the dedifferentiated ChS cell samples with a poor prognosis than in the samples with a better prognosis, and the specific inhibition of EphA2 affected the survival rate of ChS cells [186].

In addition, recent studies have shown that the expression level of FBXO22 in recurrent ChS is significantly increased, which may be related to the recurrence and progression of tumors [187]. Studies have shown that the inhibition of FBXO22 can not only reduce the proliferation and migration of ChS cells, but also promote apoptosis [187]. More interestingly, the inhibition of FBXO22 also increased the expression of PD-L1, which provides a new therapeutic strategy for immunotherapy [187]. However, not all patients with chondrosarcoma will respond to immunotherapy, so it is important to predict which patients are most likely to benefit from this treatment.

Ewing sarcoma

Ewing sarcoma (ES) is a rare, aggressive sarcoma that occurs more frequently in adolescents and young adults, ranking third among the most common primary bone tumors [188]. Current multi-agent chemotherapy and local therapies have a poor prognosis for patients with metastatic or recurrent disease, including current treatments for ES in addition to first-line chemotherapeutic agents (e.g., vincristine, doxorubicin, cyclophosphamide, etoposide, etc. [189]), gene therapy (e.g., EWSR1::FLI1 [190]), PARP inhibitors (e.g., irinotecan and temozolomide [191, 192]), and quadrivalent DR5 agonist antibodies (e.g., INBRX-109 [193]). In recent years, emerging cellular immunotherapy methods, including immune checkpoint inhibitors and CAR-T cell therapy, have attracted much attention in ES [188].

For combined macrophage-induced innate immunotherapy for ES, the researchers found that ES cells evade macrophage phagocytosis by simultaneously overexpressing CD47 and downregulating cell surface calreticulin (csCRT) [194]. The study proposes the use of a combination of CD47 blockers (magrolimab, MAG) and chemotherapeutic agents (e.g., doxorubicin, DOX) to enhance macrophage phagocytosis of ES cells, thereby inhibiting tumor growth and metastasis [194].

As with OS and ChS, EphA2 expression was significantly increased in ES compared to normal tissues, suggesting that EphA2 may be closely associated with tumor aggressive behavior [186, 195]. CAV-1 plays an important role in the angiogenesis of a variety of tumors as a key membrane transport regulator in the process of tumor angiogenesis [196]. In addition, the complex formed by EphA2 and CAV-1 can enhance the expression and release of basic fibroblast growth factor (bFGF), thereby promoting endothelial cell migration [195, 197]. Notably, the phosphorylation of EphA2 at Ser897 is closely related to tumor aggressiveness [195].

A comprehensive treatment strategy targeting EphA2 overexpression may be an effective approach for the evaluation of patients with advanced ES. Future successful use of immunotherapies in Ewing's sarcoma may rely on a combination strategy to enhance anti-tumor immunity while restoring the immunosuppressive tumor microenvironment.

Giant cell tumor of bone

Giant cell tumor of bone (GCTB) is an aggressive tumor characterized by osteolytic destruction, which usually occurs in the long metaphysis and epiphysis and spine between the ages of 20 and 40 years, with a high recurrence rate after surgical treatment, frequent local recurrence, and rare metastasis [198]. In the course of immunotherapy for GCTB, denosumab inhibits the activity of tumor cells by targeting the nuclear factor kappa B receptor activating factor ligand (RANKL), which can inhibit the activity of osteoclasts, thereby reducing the destruction of bone by GCTB cells [199,200,201].

The long-term safety and recurrence of denosumab in the treatment of GCTB are also of concern to investigators. However, Denosumab's inhibition of osteoclast activity may lead to morphological changes in GCTB cells during treatment, which may be similar to those characteristic of malignant tumor cells [202]. Denosumab has been reported to cause serious complications such as mandibular osteonecrosis, and long-term use may affect the normal development and healing process of bones [198]. In vitro studies have shown that denosumab causes an inhibitory response to tumor stromal cells rather than a cytotoxic response, which means that tumor cells may remain alive after treatment, increasing the risk of tumor recurrence after discontinuation of the drug [203]. The use of denosumab prior to curettage of tumors may increase the risk of GCTB recurrence [204]. The frequency of PD-L1 hyperexpression and CD47-SIRPα+ cell infiltration in relapsed lesions treated with denosumab was significantly higher than that in primary lesions or relapsed lesions not treated with denosumab [204]. PD-L1 and CD47-SIRPα immune checkpoint inhibitors may provide clinical benefit in patients with GCTB whose lesions have recurred after denosumab treatment [204].

In recent years, transcriptome analysis by RNA sequencing has found that the expression of Siglec-15 in GCTB tissues is high, which is significantly associated with Campanacci stage, tumor recurrence and poor prognosis [205]. Knockdown of Siglec-15 decreases the proliferation, migration, and invasion of GCTB stromal cells [205]. CXCL8 may be a gene downstream of Siglec-15 and may be associated with the CXCL2, CXCL3, CXCL5, CXCL10, CXCL11, ADORA1, BDKRB1, C5AR1, and NMU genes [205]. It is important to note that although immunotherapy methods have shown potential in laboratory studies and early clinical trials, most are still in the research and development stage, and more clinical trials are needed to verify the safety and efficacy of these treatments and determine the optimal treatment strategy.

Current methods for reshaping the BM niche of hematopoietic stem cells

The artificial niche based on biomaterials and the interactions of hematopoietic stem cells, bone marrow mesenchymal stem cells, endothelial cells, and other cells has potential clinical significance in promoting bone regeneration and treating blood diseases. The development of biomaterials has made it possible to achieve the expansion and/or differentiation of HSCs in vitro, mimicking the BM niche microenvironment by using two- or three-dimensional (3D) culture systems [206]. 2D in vitro culture revealed that the key ECM proteins and cellular ligands that promote HSC amplification are sensitive not only to biochemistry but also to the physical properties of their environment [207]. Recent studies have found that the use of poly-l-lysine (PLL)-coated biomaterials enhances the proliferation of CD34 + HSCs and promotes the differentiation of HSCs into erythrocyte progenitor cells [208]. In 3D culture systems, hydrogels and inorganic bone-like mock scaffolds are currently widely used, and co-culture of CD34 + HSCs and MSCs in BM has been shown to be effective in amplifying HSCs and secreting ECM proteins [208, 209]. HSCs proliferate higher than 2D co-cultures of 3D-MSCs, 3D-osteoblasts, and other cell types using HSCs in 3D mixed co-cultures with BMMSCs and OBs in human-derived bone scaffolds [206]. In addition, in a study on HSC differentiation, three-dimensional encapsulation of cells in pullulan (polysaccharide-based) hydrogel delayed the differentiation of HSCs into megakaryocytes and prolonged the viability and stem cell properties of HSCs [210].

Co-culture of HSCs with MSCs has been shown to be similar to native BM niche [211]. Studies have shown that encapsulation in hydrogels with low diffusivity (high collagen concentration, low Lin + BM cells) increases the total number of raw HSCs [209, 211]. The use of bioengineering methods, such as static and dynamic culture methods and microfluidic technology, not only promotes the understanding of the complex biological characteristics of HSCs in physiological and pathological states, but also brings new insights into the proliferation and bone repair of HSCs in the future. In the future, the preparation of materials for HSC amplification in BM niche will require higher ECM concentrations, higher material stiffness, uniform physical properties and molecular composition, predictable and controllable mechanical properties, and reproducible and adjustable degradation rates. In addition, the use of bioengineering methods, such as static and dynamic culture methods and microfluidic technology, not only promotes the understanding of the complex biological characteristics of HSCs in physiological and pathological states, but also brings new understanding of the proliferation and bone repair of HSCs in the future.

Conclusions

The cellular components of the endosteal and vascular BM niches interact with HSCs, and the population-specific crosstalk in the niche is regulated by chemokines, such as CXCL12, SCF, and TGFβ, as well as by intracellular Notch, Wnt, and Shh signaling pathways along with certain other factors. In the BM niche, crosstalk occurs among cells, and the niche cells control the niche function and influence each other. However, no consensus exists on how HSC interaction with the BM microenvironment enables them to perform their vital functions. Although the interactions between the numerous elements that support HSC survival, division, and differentiation processes in the ecological niche have been understood through the production of some major HSC regulatory factors by BM niche cells, there is still a lack of experimental research progress on therapeutic drugs in clinical practice. Future studies should aim to understand the different types of BM niche cells completely, determine the interaction mechanism of the cellular components that constitute the BM niche, and reveal the mechanism that regulates BM hematopoietic homeostasis. Furthermore, the molecular mechanism of HSC regulation of hematopoiesis and bone dynamic conversion balance in the BM niche must be uncovered. In the future, researchers will benefit from the widespread application of HSC-specific humanized mice, single-cell RNA sequencing, mass cytometry, whole bone sections, and in vivo animal imaging. These developments will allow the investigation of the effect and involvement of these niche cells in the occurrence and progression of tumor diseases, the elucidation of the molecular mechanisms controlling these processes, and the examination of their potential as targets for therapeutics.

Availability of data and materials

Not applicable.

Abbreviations

BM:

Bone marrow

HSCs:

Hematopoietic stem cells

ECM:

Extracellular matrix

CXCL12:

Chemokine (C-X-C motif) ligand 12

SCF:

Stem cell factor

RUNX:

Family transcription factor

BMP4:

Bone morphogenetic protein 4

VEGF:

Vascular endothelial growth factor

NGF:

Nerve growth factor

MKs:

Megakaryocytes

LEPR:

Leptin receptor

CAR cells:

CXCL12-abundant reticular cells

PDGFR:

Platelet derived growth factor receptor

PPAR-γ:

Peroxisome proliferator-activated receptor γ

TPO:

Thrombopoietin

SNS:

Sympathetic nervous system

PNS:

Parasympathetic nervous system

Ebf:

Early B cell factor

NCAD:

N-cadherin

AECs:

Arteriolar endothelial cells

SECs:

Sinusoidal endothelial cells

TGF:

Transforming growth factor

Treg cells:

Foxp3+ regulatory T cells

GFAP:

Glial fibrillary acidic protein

MBP:

Myelin basic protein

SOVs:

Small osteoblast vesicles

MOBs:

Mature osteoblasts

BMUs:

Basic multicellular units

OMs:

Osteomacs

OPG:

Osteoprotegerin

SOX9:

Sex-determining region of Y chromosome (SRY)-box transcription factor 9

OS:

Osteosarcoma

ChS:

Osteochondroma

ES:

Ewing sarcoma

GCTB:

Giant cell tumor of bone

TME:

Tumor microenvironment

TAMs:

Tumor-associated macrophages

References

  1. Fang H, Deng Z, Liu J, Chen S, Deng Z, Li W. The mechanism of bone remodeling after bone aging. Clin Interv Aging. 2022;17:405–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Yao D, Huang L, Ke J, Zhang M, Xiao Q, Zhu X. Bone metabolism regulation: Implications for the treatment of bone diseases. Biomed Pharmacother. 2020;129: 110494.

    Article  CAS  PubMed  Google Scholar 

  3. Lee-Thedieck C, Schertl P, Klein G. The extracellular matrix of hematopoietic stem cell niches. Adv Drug Deliv Rev. 2022;181: 114069.

    Article  CAS  PubMed  Google Scholar 

  4. Pinho S, Frenette PS. Haematopoietic stem cell activity and interactions with the niche. Nat Rev Mol Cell Biol. 2019;20:303–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Crane GM, Jeffery E, Morrison SJ. Adult haematopoietic stem cell niches. Nat Rev Immunol. 2017;17:573–90.

    Article  CAS  PubMed  Google Scholar 

  6. Aoki K, Kurashige M, Ichii M, Higaki K, Sugiyama T, Kaito T, Ando W, Sugano N, Sakai T, Shibayama H, et al. Identification of CXCL12-abundant reticular cells in human adult bone marrow. Br J Haematol. 2021;193:659–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Morrison SJ, Scadden DT. The bone marrow niche for haematopoietic stem cells. Nature. 2014;505:327–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang X, Hou L, Li F, Zhang W, Wu C, Xiang L, Li J, Zhou L, Wang X, Xiang Y, et al. Piezo1-mediated mechanosensation in bone marrow macrophages promotes vascular niche regeneration after irradiation injury. Theranostics. 2022;12:1621–38.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Shen B, Tasdogan A, Ubellacker JM, Zhang J, Nosyreva ED, Du L, Murphy MM, Hu S, Yi Y, Kara N, et al. A mechanosensitive peri-arteriolar niche for osteogenesis and lymphopoiesis. Nature. 2021;591:438–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Salhotra A, Shah HN, Levi B, Longaker MT. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol. 2020;21:696–711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sukhnanan K, Ross JR, Chao NJ, Chen BJ. Endothelial cell derived extracellular vesicles and hematopoiesis. Radiat Res. 2024;202(2):215–26.

    Article  CAS  PubMed  Google Scholar 

  12. Ho NP, Takizawa H. Inflammation regulates haematopoietic stem cells and their niche. Int J Mol Sci. 2022;23(3):1125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Aerts-Kaya F, Ulum B, Mammadova A, Kose S, Aydin G, Korkusuz P, Uckan-Cetinkaya D. Neurological regulation of the bone marrow niche. Adv Exp Med Biol. 2020;1212:127–53.

    Article  CAS  PubMed  Google Scholar 

  14. Owen-Woods C, Kusumbe A. Fundamentals of bone vasculature: Specialization, interactions and functions. Semin Cell Dev Biol. 2022;123:36–47.

    Article  CAS  PubMed  Google Scholar 

  15. Ghosh J, Koussa RE, Mohamad SF, Liu J, Kacena MA, Srour EF. Cellular components of the hematopoietic niche and their regulation of hematopoietic stem cell function. Curr Opin Hematol. 2021;28:243–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ambrosi TH, Marecic O, McArdle A, Sinha R, Gulati GS, Tong X, Wang Y, Steininger HM, Hoover MY, Koepke LS, et al. Aged skeletal stem cells generate an inflammatory degenerative niche. Nature. 2021;597:256–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Deng Z, Zhang Q, Zhao Z, Li Y, Chen X, Lin Z, Deng Z, Liu J, Duan L, Wang D, Li W. Crosstalk between immune cells and bone cells or chondrocytes. Int Immunopharmacol. 2021;101: 108179.

    Article  CAS  PubMed  Google Scholar 

  18. Dennis JE, Charbord P. Origin and differentiation of human and murine stroma. Stem Cells. 2002;20:205–14.

    Article  CAS  PubMed  Google Scholar 

  19. Bhat S, Viswanathan P, Chandanala S, Prasanna SJ, Seetharam RN. Expansion and characterization of bone marrow derived human mesenchymal stromal cells in serum-free conditions. Sci Rep. 2021;11:3403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Drela K, Stanaszek L, Snioch K, Kuczynska Z, Wrobel M, Sarzynska S, Legosz P, Maldyk P, Lukomska B. Bone marrow-derived from the human femoral shaft as a new source of mesenchymal stem/stromal cells: an alternative cell material for banking and clinical transplantation. Stem Cell Res Ther. 2020;11:262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wang YH, Zhao CZ, Wang RY, Du QX, Liu JY, Pan J. The crosstalk between macrophages and bone marrow mesenchymal stem cells in bone healing. Stem Cell Res Ther. 2022;13:511.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zhao M, Tao F, Venkatraman A, Li Z, Smith SE, Unruh J, Chen S, Ward C, Qian P, Perry JM, et al. N-cadherin-expressing bone and marrow stromal progenitor cells maintain reserve hematopoietic stem cells. Cell Rep. 2019;26:652-669.e656.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Prasad P, Cancelas JA. From marrow to bone and fat: Exploring the multifaceted roles of leptin receptor positive bone marrow mesenchymal stromal cells. Cells. 2024;13:910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Crisan M, Kartalaei PS, Vink CS, Yamada-Inagawa T, Bollerot K, van Ijcken W, van der Linden R, de Sousa LSMC, Monteiro R, Mummery C, Dzierzak E. Corrigendum: BMP signalling differentially regulates distinct haematopoietic stem cell types. Nat Commun. 2015;6:8793.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Crisan M, Solaimani KP, Neagu A, Karkanpouna S, Yamada-Inagawa T, Purini C, Vink CS, van der Linden R, van Ijcken W, Chuva DSLSM, et al. BMP and hedgehog regulate distinct AGM hematopoietic stem cells ex vivo. Stem Cell Reports. 2016;6:383–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Pinho S, Lacombe J, Hanoun M, Mizoguchi T, Bruns I, Kunisaki Y, Frenette PS. PDGFRalpha and CD51 mark human nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. J Exp Med. 2013;210:1351–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Asada N, Kunisaki Y, Pierce H, Wang Z, Fernandez NF, Birbrair A, Ma’Ayan A, Frenette PS. Differential cytokine contributions of perivascular haematopoietic stem cell niches. Nat Cell Biol. 2017;19:214–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Galan-Diez M, Kousteni S. A bone marrow niche-derived molecular switch between osteogenesis and hematopoiesis. Genes Dev. 2018;32:324–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Baccin C, Al-Sabah J, Velten L, Helbling PM, Grunschlager F, Hernandez-Malmierca P, Nombela-Arrieta C, Steinmetz LM, Trumpp A, Haas S. Combined single-cell and spatial transcriptomics reveal the molecular, cellular and spatial bone marrow niche organization. Nat Cell Biol. 2020;22:38–48.

    Article  CAS  PubMed  Google Scholar 

  30. Sugiyama T, Omatsu Y, Nagasawa T. Niches for hematopoietic stem cells and immune cell progenitors. Int Immunol. 2019;31:5–11.

    Article  CAS  PubMed  Google Scholar 

  31. Omatsu Y, Aiba S, Maeta T, Higaki K, Aoki K, Watanabe H, Kondoh G, Nishimura R, Takeda S, Chung U-I, Nagasawa T. Runx1 and Runx2 inhibit fibrotic conversion of cellular niches for hematopoietic stem cells. Nat Commun. 2022;13:2654–2654.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yen YT, Chien M, Wu PY, Hung SC. PP2A in LepR+ mesenchymal stem cells contributes to embryonic and postnatal endochondral ossification through Runx2 dephosphorylation. Commun Biol. 2021;4:658.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kadomatsu T, Oike Y. Roles of angiopoietin-like proteins in regulation of stem cell activity. J Biochem. 2019;165:309–15.

    Article  CAS  PubMed  Google Scholar 

  34. Kandarakov O, Belyavsky A, Semenova E. Bone marrow niches of hematopoietic stem and progenitor cells. Int J Mol Sci. 2022;23:4462.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sharma M, Ross C, Srivastava S. Ally to adversary: mesenchymal stem cells and their transformation in leukaemia. Cancer Cell Int. 2019;19:139.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Kunisaki Y, Bruns I, Scheiermann C, Ahmed J, Pinho S, Zhang D, Mizoguchi T, Wei Q, Lucas D, Ito K, et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature. 2013;502:637–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chitteti BR, Cheng YH, Streicher DA, Rodriguez-Rodriguez S, Carlesso N, Srour EF, Kacena MA. Osteoblast lineage cells expressing high levels of Runx2 enhance hematopoietic progenitor cell proliferation and function. J Cell Biochem. 2010;111:284–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lu YF, Cahan P, Ross S, Sahalie J, Sousa PM, Hadland BK, Cai W, Serrao E, Engelman AN, Bernstein ID, Daley GQ. Engineered murine HSCs reconstitute multi-lineage hematopoiesis and adaptive immunity. Cell Rep. 2016;17:3178–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sumii Y, Kondo T, Ikegawa S, Fukumi T, Iwamoto M, Nishimura MF, Sugiura H, Sando Y, Nakamura M, Meguri Y, et al. Hematopoietic stem cell-derived Tregs are essential for maintaining favorable B cell lymphopoiesis following posttransplant cyclophosphamide. JCI Insight. 2023;8: e162180.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Seike M, Omatsu Y, Watanabe H, Kondoh G, Nagasawa T. Stem cell niche-specific Ebf3 maintains the bone marrow cavity. Genes Dev. 2018;32:359–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Deng Z, Li W, Xu J, Yu M, Li D, Tan Q, Wang D, Chen L, Wang L. ClC-3chloride channels are involved in estradiol regulation of bone formation by MC3T3-E1 osteoblasts. J Cell Biochem. 2019;120:8366–75.

    Article  CAS  PubMed  Google Scholar 

  42. Kajiume T, Kawahara Y, Yuge L, Kobayashi M. Osteoblastic adherence regulates hematopoietic stem cell self-renewal and differentiation: a conceptional in vitro and in vivo study. Stem Cell Investig. 2021;8:21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Haylock DN, Nilsson SK. Osteopontin: a bridge between bone and blood. Br J Haematol. 2006;134:467–74.

    Article  CAS  PubMed  Google Scholar 

  44. Sugimura R, He XC, Venkatraman A, Arai F, Box A, Semerad C, Haug JS, Peng L, Zhong XB, Suda T, Li L. Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche. Cell. 2012;150:351–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Lee S, Zhan H. Deciphering the differential impact of thrombopoietin/MPL signaling on hematopoietic stem/progenitor cell function in bone marrow and spleen. Stem Cell Reports. 2024;19:211–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tikhonova AN, Dolgalev I, Hu H, Sivaraj KK, Hoxha E, Cuesta-Dominguez A, Pinho S, Akhmetzyanova I, Gao J, Witkowski M, et al. The bone marrow microenvironment at single-cell resolution. Nature. 2019;569:222–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Schreck C, Istvanffy R, Ziegenhain C, Sippenauer T, Ruf F, Henkel L, Gartner F, Vieth B, Florian MC, Mende N, et al. Niche WNT5A regulates the actin cytoskeleton during regeneration of hematopoietic stem cells. J Exp Med. 2017;214:165–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sacma M, Pospiech J, Bogeska R, de Back W, Mallm JP, Sakk V, Soller K, Marka G, Vollmer A, Karns R, et al. Haematopoietic stem cells in perisinusoidal niches are protected from ageing. Nat Cell Biol. 2019;21:1309–20.

    Article  CAS  PubMed  Google Scholar 

  49. Deng J, Tan Y, Xu Z, Wang H. Advances in hematopoietic stem cells ex vivo expansion associated with bone marrow niche. Ann Hematol. 2024. https://doi.org/10.1007/s00277-024-05773-1.

    Article  PubMed  Google Scholar 

  50. Rafii S, Shapiro F, Pettengell R, Ferris B, Nachman RL, Moore MA, Asch AS. Human bone marrow microvascular endothelial cells support long-term proliferation and differentiation of myeloid and megakaryocytic progenitors. Blood. 1995;86:3353–63.

    Article  CAS  PubMed  Google Scholar 

  51. Man Y, Yao X, Yang T, Wang Y. Hematopoietic stem cell niche during homeostasis, malignancy, and bone marrow transplantation. Front Cell Dev Biol. 2021;9: 621214.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Hooper AT, Butler JM, Nolan DJ, Kranz A, Iida K, Kobayashi M, Kopp HG, Shido K, Petit I, Yanger K, et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell. 2009;4:263–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Butler JM, Nolan DJ, Vertes EL, Varnum-Finney B, Kobayashi H, Hooper AT, Seandel M, Shido K, White IA, Kobayashi M, et al. Endothelial cells are essential for the self-renewal and repopulation of Notch-dependent hematopoietic stem cells. Cell Stem Cell. 2010;6:251–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Xu C, Gao X, Wei Q, Nakahara F, Zimmerman SE, Mar J, Frenette PS. Stem cell factor is selectively secreted by arterial endothelial cells in bone marrow. Nat Commun. 2018;9:2449.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Kara N, Xue Y, Zhao Z, Murphy MM, Comazzetto S, Lesser A, Du L, Morrison SJ. Endothelial and Leptin Receptor(+) cells promote the maintenance of stem cells and hematopoiesis in early postnatal murine bone marrow. Dev Cell. 2023;58:348-360.e346.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121:1109–21.

    Article  CAS  PubMed  Google Scholar 

  57. Barbier V, Erbani J, Fiveash C, Davies JM, Tay J, Tallack MR, Lowe J, Magnani JL, Pattabiraman DR, Perkins AC, et al. Endothelial E-selectin inhibition improves acute myeloid leukaemia therapy by disrupting vascular niche-mediated chemoresistance. Nat Commun. 2020;11:2042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Romo-Gonzalez M, Ijurko C, Hernandez-Hernandez A. Reactive oxygen species and metabolism in leukemia: A dangerous liaison. Front Immunol. 2022;13: 889875.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bruschi M, Biancucci F, Masini S, Piacente F, Ligi D, Bartoccini F, Antonelli A, Mannello F, Bruzzone S, Menotta M, et al. The influence of redox modulation on hypoxic endothelial cell metabolic and proteomic profiles through a small thiol-based compound tuning glutathione and thioredoxin systems. BioFactors. 2023;49:1205–22.

    Article  CAS  PubMed  Google Scholar 

  60. Lampreia FP, Carmelo JG, Anjos-Afonso F. Notch signaling in the regulation of hematopoietic stem cell. Curr Stem Cell Rep. 2017;3:202–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Poulos MG, Guo P, Kofler NM, Pinho S, Gutkin MC, Tikhonova A, Aifantis I, Frenette PS, Kitajewski J, Rafii S, Butler JM. Endothelial Jagged-1 is necessary for homeostatic and regenerative hematopoiesis. Cell Rep. 2013;4:1022–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Plein A, Fantin A, Denti L, Pollard JW, Ruhrberg C. Erythro-myeloid progenitors contribute endothelial cells to blood vessels. Nature. 2018;562:223–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Cary L, Noutai D, Salber R, Fadiyimu O, Gross A, Almeida-Porada G, Kidane Y, Whitnall M. Bone marrow endothelial cells influence function and phenotype of hematopoietic stem and progenitor cells after mixed neutron/gamma radiation. Int J Mol Sci. 2019;20:1795.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Nakamura-Ishizu A, Takubo K, Fujioka M, Suda T. Megakaryocytes are essential for HSC quiescence through the production of thrombopoietin. Biochem Biophys Res Commun. 2014;454:353–7.

    Article  CAS  PubMed  Google Scholar 

  65. Bozhilov YK, Hsu I, Brown EJ, Wilkinson AC. In vitro human haematopoietic stem cell expansion and differentiation. Cells. 2023;12:896.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Bruns I, Lucas D, Pinho S, Ahmed J, Lambert MP, Kunisaki Y, Scheiermann C, Schiff L, Poncz M, Bergman A, Frenette PS. Megakaryocytes regulate hematopoietic stem cell quiescence through CXCL4 secretion. Nat Med. 2014;20:1315–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Norozi F, Shahrabi S, Hajizamani S, Saki N. Regulatory role of megakaryocytes on hematopoietic stem cells quiescence by CXCL4/PF4 in bone marrow niche. Leuk Res. 2016;48:107–12.

    Article  CAS  PubMed  Google Scholar 

  68. Zhao M, Perry JM, Marshall H, Venkatraman A, Qian P, He XC, Ahamed J, Li L. Megakaryocytes maintain homeostatic quiescence and promote post-injury regeneration of hematopoietic stem cells. Nat Med. 2014;20:1321–6.

    Article  CAS  PubMed  Google Scholar 

  69. Tang Y, Hu M, Xu Y, Chen F, Chen S, Chen M, Qi Y, Shen M, Wang C, Lu Y, et al. Megakaryocytes promote bone formation through coupling osteogenesis with angiogenesis by secreting TGF-beta1. Theranostics. 2020;10:2229–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Gostynska S, Venkatesan T, Subramani K, Cortez B, Robertson A, Subrahmanian S, Dube P, Ahamed J. Megakaryocyte/platelet-derived TGF-beta1 inhibits megakaryopoiesis in bone marrow by regulating thrombopoietin production in liver. Blood Adv. 2022;6:3321–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Tsutsumi N, Masoumi Z, James SC, Tucker JA, Winkelmann H, Grey W, Picton LK, Moss L, Wilson SC, Caveney NA, et al. Structure of the thrombopoietin-MPL receptor complex is a blueprint for biasing hematopoiesis. Cell. 2023;186:4189-4203.e4122.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Cui L, Moraga I, Lerbs T, Van Neste C, Wilmes S, Tsutsumi N, Trotman-Grant AC, Gakovic M, Andrews S, Gotlib J, et al. Tuning MPL signaling to influence hematopoietic stem cell differentiation and inhibit essential thrombocythemia progenitors. Proc Natl Acad Sci U S A. 2021;118: e2017849118.

    Article  CAS  PubMed  Google Scholar 

  73. Nakamura-Ishizu A, Takubo K, Kobayashi H, Suzuki-Inoue K, Suda T. Correction: CLEC-2 in megakaryocytes is critical for maintenance of hematopoietic stem cells in the bone marrow. J Exp Med. 2015;212:2323.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Ludin A, Itkin T, Gur-Cohen S, Mildner A, Shezen E, Golan K, Kollet O, Kalinkovich A, Porat Z, D’Uva G, et al. Monocytes-macrophages that express alpha-smooth muscle actin preserve primitive hematopoietic cells in the bone marrow. Nat Immunol. 2012;13:1072–82.

    Article  CAS  PubMed  Google Scholar 

  75. Acar M, Kocherlakota KS, Murphy MM, Peyer JG, Oguro H, Inra CN, Jaiyeola C, Zhao Z, Luby-Phelps K, Morrison SJ. Deep imaging of bone marrow shows non-dividing stem cells are mainly perisinusoidal. Nature. 2015;526:126–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Peterova E, Podmolikova L, Rezacova M, Mrkvicova A. Fibroblast growth factor-1 suppresses TGF-beta-mediated myofibroblastic differentiation of rat hepatic stellate cells. Acta Medica (Hradec Kralove). 2016;59:124–32.

    Article  CAS  PubMed  Google Scholar 

  77. Dutta P, Hoyer FF, Grigoryeva LS, Sager HB, Leuschner F, Courties G, Borodovsky A, Novobrantseva T, Ruda VM, Fitzgerald K, et al. Macrophages retain hematopoietic stem cells in the spleen via VCAM-1. J Exp Med. 2015;212:497–512.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. May A, Forrester LM. The erythroblastic island niche: modeling in health, stress, and disease. Exp Hematol. 2020;91:10–21.

    Article  CAS  PubMed  Google Scholar 

  79. Yang C, Endoh M, Tan DQ, Nakamura-Ishizu A, Takihara Y, Matsumura T, Suda T. Mitochondria transfer from early stages of erythroblasts to their macrophage niche via tunnelling nanotubes. Br J Haematol. 2021;193:1260–74.

    Article  CAS  PubMed  Google Scholar 

  80. Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, Poulton IJ, van Rooijen N, Alexander KA, Raggatt LJ, Levesque JP. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116:4815–28.

    Article  CAS  PubMed  Google Scholar 

  81. Christopher MJ, Rao M, Liu F, Woloszynek JR, Link DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J Exp Med. 2011;208:251–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Lucas D, Bruns I, Battista M, Mendez-Ferrer S, Magnon C, Kunisaki Y, Frenette PS. Norepinephrine reuptake inhibition promotes mobilization in mice: potential impact to rescue low stem cell yields. Blood. 2012;119:3962–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Putra A, Alif I, Hamra N, Santosa O, Kustiyah AR, Muhar AM, Lukman K. MSC-released TGF-beta regulate alpha-SMA expression of myofibroblast during wound healing. J Stem Cells Regen Med. 2020;16:73–9.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Pan Y, Cao S, Tang J, Arroyo JP, Terker AS, Wang Y, Niu A, Fan X, Wang S, Zhang Y, et al. Cyclooxygenase-2 in adipose tissue macrophages limits adipose tissue dysfunction in obese mice. J Clin Invest. 2022;132: e152391.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Hur J, Choi JI, Lee H, Nham P, Kim TW, Chae CW, Yun JY, Kang JA, Kang J, Lee SE, et al. CD82/KAI1 maintains the dormancy of long-term hematopoietic stem cells through interaction with DARC-expressing macrophages. Cell Stem Cell. 2016;18:508–21.

    Article  CAS  PubMed  Google Scholar 

  86. Ding L, Morrison SJ. Haematopoietic stem cells and early lymphoid progenitors occupy distinct bone marrow niches. Nature. 2013;495:231–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Fujisaki J, Wu J, Carlson AL, Silberstein L, Putheti P, Larocca R, Gao W, Saito TI, Lo CC, Tsuyuzaki H, et al. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature. 2011;474:216–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Lucas D. Leukocyte trafficking and regulation of murine hematopoietic stem cells and their niches. Front Immunol. 2019;10:387–387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Camacho V, Matkins VR, Patel SB, Lever JM, Yang Z, Ying L, Landuyt AE, Dean EC, George JF, Yang H, et al. Bone marrow Tregs mediate stromal cell function and support hematopoiesis via IL-10. JCI Insight. 2020;5: e135681.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Hirata Y, Furuhashi K, Ishii H, Li HW, Pinho S, Ding L, Robson SC, Frenette PS, Fujisaki J. CD150(high) bone marrow tregs maintain hematopoietic stem cell quiescence and immune privilege via adenosine. Cell Stem Cell. 2018;22:445-453.e445.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Matsushita Y, Nagata M, Kozloff KM, Welch JD, Mizuhashi K, Tokavanich N, Hallett SA, Link DC, Nagasawa T, Ono W, Ono N. A Wnt-mediated transformation of the bone marrow stromal cell identity orchestrates skeletal regeneration. Nat Commun. 2020;11:332.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Kakiuchi M, Hirata Y, Robson SC, Fujisaki J. Paradoxical regulation of allogeneic bone marrow engraftment and immune privilege by mesenchymal cells and adenosine. Transplant Cell Ther. 2021;27:92.e91-92.e95.

    Google Scholar 

  93. Schloss MJ, Hulsmans M, Rohde D, Lee IH, Severe N, Foy BH, Pulous FE, Zhang S, Kokkaliaris KD, Frodermann V, et al. Author Correction: B lymphocyte-derived acetylcholine limits steady-state and emergency hematopoiesis. Nat Immunol. 2022;23:1285.

    Article  PubMed  Google Scholar 

  94. Guerra DAP, Paiva AE, Sena IFG, Azevedo PO, Batista ML Jr, Mintz A, Birbrair A. Adipocytes role in the bone marrow niche. Cytometry A. 2018;93:167–71.

    Article  PubMed  Google Scholar 

  95. He N, Liu M, Wu Y. Adipose tissue and hematopoiesis: Friend or foe? J Clin Lab Anal. 2023;37: e24872.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Tratwal J, Rojas-Sutterlin S, Bataclan C, Blum S, Naveiras O. Bone marrow adiposity and the hematopoietic niche: A historical perspective of reciprocity, heterogeneity, and lineage commitment. Best Pract Res Clin Endocrinol Metab. 2021;35: 101564.

    Article  CAS  PubMed  Google Scholar 

  97. Liu X, Gu Y, Kumar S, Amin S, Guo Q, Wang J, Fang CL, Cao X, Wan M. Oxylipin-PPARgamma-initiated adipocyte senescence propagates secondary senescence in the bone marrow. Cell Metab. 2023;35:667-684.e666.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wilson A, Fu H, Schiffrin M, Winkler C, Koufany M, Jouzeau JY, Bonnet N, Gilardi F, Renevey F, Luther SA, et al. Lack of adipocytes alters hematopoiesis in lipodystrophic mice. Front Immunol. 2018;9:2573.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Ferland-McCollough D, Maselli D, Spinetti G, Sambataro M, Sullivan N, Blom A, Madeddu P. MCP-1 feedback loop between adipocytes and mesenchymal stromal cells causes fat accumulation and contributes to hematopoietic stem cell rarefaction in the bone marrow of patients with diabetes. Diabetes. 2018;67:1380–94.

    Article  CAS  PubMed  Google Scholar 

  100. Meacham CE, Jeffery EC, Burgess RJ, Sivakumar CD, Arora MA, Stanley AM, Colby EM, Crane GM, Zhao Z, Morrison SJ. Adiponectin receptors sustain haematopoietic stem cells throughout adulthood by protecting them from inflammation. Nat Cell Biol. 2022;24:697–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. DiMascio L, Voermans C, Uqoezwa M, Duncan A, Lu D, Wu J, Sankar U, Reya T. Identification of adiponectin as a novel hemopoietic stem cell growth factor. J Immunol. 2007;178:3511–20.

    Article  CAS  PubMed  Google Scholar 

  102. Li J, Chen T, Huang X, Zhao Y, Wang B, Yin Y, Cui Y, Zhao Y, Zhang R, Wang X, et al. Substrate-independent immunomodulatory characteristics of mesenchymal stem cells in three-dimensional culture. PLoS ONE. 2018;13: e0206811.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Yue R, Zhou BO, Shimada IS, Zhao Z, Morrison SJ. Leptin receptor promotes adipogenesis and reduces osteogenesis by regulating mesenchymal stromal cells in adult bone marrow. Cell Stem Cell. 2016;18:782–96.

    Article  CAS  PubMed  Google Scholar 

  104. Mattiucci D, Maurizi G, Izzi V, Cenci L, Ciarlantini M, Mancini S, Mensa E, Pascarella R, Vivarelli M, Olivieri A, et al. Bone marrow adipocytes support hematopoietic stem cell survival. J Cell Physiol. 2018;233:1500–11.

    Article  CAS  PubMed  Google Scholar 

  105. Zhou BO, Yu H, Yue R, Zhao Z, Rios JJ, Naveiras O, Morrison SJ. Bone marrow adipocytes promote the regeneration of stem cells and haematopoiesis by secreting SCF. Nat Cell Biol. 2017;19:891–903.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Hanoun M, Maryanovich M, Arnal-Estape A, Frenette PS. Neural regulation of hematopoiesis, inflammation, and cancer. Neuron. 2015;86:360–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Jung WC, Levesque JP, Ruitenberg MJ. It takes nerve to fight back: The significance of neural innervation of the bone marrow and spleen for immune function. Semin Cell Dev Biol. 2017;61:60–70.

    Article  CAS  PubMed  Google Scholar 

  108. Christodoulou C, Spencer JA, Yeh SA, Turcotte R, Kokkaliaris KD, Panero R, Ramos A, Guo G, Seyedhassantehrani N, Esipova TV, et al. Live-animal imaging of native haematopoietic stem and progenitor cells. Nature. 2020;578:278–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452:442–7.

    Article  CAS  PubMed  Google Scholar 

  110. Maryanovich M, Zahalka AH, Pierce H, Pinho S, Nakahara F, Asada N, Wei Q, Wang X, Ciero P, Xu J, et al. Author Correction: Adrenergic nerve degeneration in bone marrow drives aging of the hematopoietic stem cell niche. Nat Med. 2019;25:701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Heidt T, Sager HB, Courties G, Dutta P, Iwamoto Y, Zaltsman A, von Zur MC, Bode C, Fricchione GL, Denninger J, et al. Chronic variable stress activates hematopoietic stem cells. Nat Med. 2014;20:754–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Khan E, Shrestha AK, Elkhooly M, Wilson H, Ebbert M, Srivastava S, Wen S, Rollins S, Sriwastava S. CNS and PNS manifestation in immune checkpoint inhibitors: A systematic review. J Neurol Sci. 2022;432: 120089.

    Article  CAS  PubMed  Google Scholar 

  113. Shindo Y, Nakamura HM, Nakai J, Wakamori M, Nakamura T. A parasympathetic neurotransmitter induces myoepithelial cell differentiation during salivary gland development. Exp Cell Res. 2022;416: 113137.

    Article  CAS  PubMed  Google Scholar 

  114. Bajayo A, Bar A, Denes A, Bachar M, Kram V, Attar-Namdar M, Zallone A, Kovacs KJ, Yirmiya R, Bab I. Skeletal parasympathetic innervation communicates central IL-1 signals regulating bone mass accrual. Proc Natl Acad Sci U S A. 2012;109:15455–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Garcia-Garcia A, Korn C, Garcia-Fernandez M, Domingues O, Villadiego J, Martin-Perez D, Isern J, Bejarano-Garcia JA, Zimmer J, Perez-Simon JA, et al. Dual cholinergic signals regulate daily migration of hematopoietic stem cells and leukocytes. Blood. 2019;133:224–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Fielding C, Garcia-Garcia A, Korn C, Gadomski S, Fang Z, Reguera JL, Perez-Simon JA, Gottgens B, Mendez-Ferrer S. Cholinergic signals preserve haematopoietic stem cell quiescence during regenerative haematopoiesis. Nat Commun. 2022;13:543.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Pierce H, Zhang D, Magnon C, Lucas D, Christin JR, Huggins M, Schwartz GJ, Frenette PS. Cholinergic signals from the CNS regulate G-CSF-mediated HSC mobilization from bone marrow via a glucocorticoid signaling relay. Cell Stem Cell. 2017;20:648-658.e644.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Nocera G, Jacob C. Mechanisms of Schwann cell plasticity involved in peripheral nerve repair after injury. Cell Mol Life Sci. 2020;77:3977–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Louit A, Beaudet MJ, Pepin R, Berthod F. Differentiation of human induced pluripotent stem cells into mature and myelinating Schwann cells. Tissue Eng Part C Methods. 2023;29:134–43.

    Article  CAS  PubMed  Google Scholar 

  120. Kastriti ME, Faure L, Von Ahsen D, Bouderlique TG, Bostrom J, Solovieva T, Jackson C, Bronner M, Meijer D, Hadjab S, et al. Schwann cell precursors represent a neural crest-like state with biased multipotency. EMBO J. 2022;41: e108780.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Ding Z, Jiang M, Qian J, Gu D, Bai H, Cai M, Yao D. Role of transforming growth factor-beta in peripheral nerve regeneration. Neural Regen Res. 2024;19:380–6.

    Article  CAS  PubMed  Google Scholar 

  122. Gordon T. Peripheral nerve regeneration and muscle reinnervation. Int J Mol Sci. 2020;21:8652.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Kataria H, Alizadeh A, Karimi-Abdolrezaee S. Neuregulin-1/ErbB network: An emerging modulator of nervous system injury and repair. Prog Neurobiol. 2019;180: 101643.

    Article  CAS  PubMed  Google Scholar 

  124. Maestroni GJM. Adrenergic modulation of hematopoiesis. J Neuroimmune Pharmacol. 2020;15:82–92.

    Article  PubMed  Google Scholar 

  125. Uenaka M, Yamashita E, Kikuta J, Morimoto A, Ao T, Mizuno H, Furuya M, Hasegawa T, Tsukazaki H, Sudo T, et al. Osteoblast-derived vesicles induce a switch from bone-formation to bone-resorption in vivo. Nat Commun. 2022;13:1066–1066.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Kim JM, Lin C, Stavre Z, Greenblatt MB, Shim JH. Osteoblast-osteoclast communication and bone homeostasis. Cells. 2020;9:2073.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Zhang Y, Luo G, Yu X. Cellular communication in bone homeostasis and the related anti-osteoporotic drug development. Curr Med Chem. 2020;27:1151–69.

    Article  CAS  PubMed  Google Scholar 

  128. Miyamoto K, Yoshida S, Kawasumi M, Hashimoto K, Kimura T, Sato Y, Kobayashi T, Miyauchi Y, Hoshi H, Iwasaki R, et al. Osteoclasts are dispensable for hematopoietic stem cell maintenance and mobilization. J Exp Med. 2011;208:2175–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Lucas D. Structural organization of the bone marrow and its role in hematopoiesis. Curr Opin Hematol. 2021;28:36–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Tamma R, Zallone A. Osteoblast and osteoclast crosstalks: from OAF to Ephrin. Inflamm Allergy Drug Targets. 2012;11:196–200.

    Article  CAS  PubMed  Google Scholar 

  131. Matsuo K, Irie N. Osteoclast-osteoblast communication. Arch Biochem Biophys. 2008;473:201–9.

    Article  CAS  PubMed  Google Scholar 

  132. Nara Y, Kitaura H, Ogawa S, Shen WR, Qi J, Ohori F, Noguchi T, Marahleh A, Pramusita A, Kinjo R, Mizoguchi I. Anti-c-fms antibody prevents osteoclast formation and bone resorption in co-culture of osteoblasts and osteoclast precursors in vitro and in ovariectomized mice. Int J Mol Sci. 2020;21:6120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  134. Kim JH, Kim N. Bone cell communication factors provide a new therapeutic strategy for osteoporosis. Chonnam Med J. 2020;56:94–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Zhou J, Zhu Y, Ai D, Zhou M, Li H, Fu Y, Song J. Low-intensity pulsed ultrasound regulates osteoblast-osteoclast crosstalk via EphrinB2/EphB4 signaling for orthodontic alveolar bone remodeling. Front Bioeng Biotechnol. 2023;11:1192720.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Ge YW, Feng K, Liu XL, Chen HF, Sun ZY, Wang CF, Liu ZQ, Wang HW, Zhang JW, Yu DG, Mao YQ. The recombinant protein EphB4-Fc changes the Ti particle-mediated imbalance of OPG/RANKL via EphrinB2/EphB4 signaling pathway and inhibits the release of proinflammatory factors in vivo. Oxid Med Cell Longev. 2020;2020:1404915.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Lee YS, Kwak MK, Moon SA, Choi YJ, Baek JE, Park SY, Kim BJ, Lee SH, Koh JM. Regulation of bone metabolism by megakaryocytes in a paracrine manner. Sci Rep. 2020;10:2277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Eleniste PP, Patel V, Posritong S, Zero O, Largura H, Cheng YH, Himes ER, Hamilton M, Ekwealor JTB, Kacena MA, Bruzzaniti A. Pyk2 and megakaryocytes regulate osteoblast differentiation and migration via distinct and overlapping mechanisms. J Cell Biochem. 2016;117:1396–406.

    Article  CAS  PubMed  Google Scholar 

  139. Gangat N, Tefferi A. Myelofibrosis biology and contemporary management. Br J Haematol. 2020;191:152–70.

    Article  CAS  PubMed  Google Scholar 

  140. Noetzli LJ, French SL, Machlus KR. New insights into the differentiation of megakaryocytes from hematopoietic progenitors. Arterioscler Thromb Vasc Biol. 2019;39:1288–300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Cheng YH, Chitteti BR, Streicher DA, Morgan JA, Rodriguez-Rodriguez S, Carlesso N, Srour EF, Kacena MA. Impact of maturational status on the ability of osteoblasts to enhance the hematopoietic function of stem and progenitor cells. J Bone Miner Res. 2011;26:1111–21.

    Article  CAS  PubMed  Google Scholar 

  142. Ciovacco WA, Cheng YH, Horowitz MC, Kacena MA. Immature and mature megakaryocytes enhance osteoblast proliferation and inhibit osteoclast formation. J Cell Biochem. 2010;109:774–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Maupin KA, Himes ER, Plett AP, Chua HL, Singh P, Ghosh J, Mohamad SF, Abeysekera I, Fisher A, Sampson C, et al. Aging negatively impacts the ability of megakaryocytes to stimulate osteoblast proliferation and bone mass. Bone. 2019;127:452–9.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Olson TS, Caselli A, Otsuru S, Hofmann TJ, Williams R, Paolucci P, Dominici M, Horwitz EM. Megakaryocytes promote murine osteoblastic HSC niche expansion and stem cell engraftment after radioablative conditioning. Blood. 2013;121:5238–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Frisch BJ, Hoffman CM, Latchney SE, LaMere MW, Myers J, Ashton J, Li AJ, Saunders JN, Palis J, Perkins AS, et al. Aged marrow macrophages expand platelet-biased hematopoietic stem cells via Interleukin1B. JCI Insight. 2019;5: e124213.

    Article  PubMed  Google Scholar 

  146. Kuribayashi W, Oshima M, Itokawa N, Koide S, Nakajima-Takagi Y, Yamashita M, Yamazaki S, Rahmutulla B, Miura F, Ito T, et al. Limited rejuvenation of aged hematopoietic stem cells in young bone marrow niche. J Exp Med. 2021;218: e20192283.

    Article  CAS  PubMed  Google Scholar 

  147. Croes M, Oner FC, van Neerven D, Sabir E, Kruyt MC, Blokhuis TJ, Dhert WJA, Alblas J. Proinflammatory T cells and IL-17 stimulate osteoblast differentiation. Bone. 2016;84:262–70.

    Article  CAS  PubMed  Google Scholar 

  148. Cifuentes-Mendiola SE, Solis-Suarez DL, Martinez-Davalos A, Godinez-Victoria M, Garcia-Hernandez AL. CD4(+) T-cell activation of bone marrow causes bone fragility and insulin resistance in type 2 diabetes. Bone. 2022;155: 116292.

    Article  CAS  PubMed  Google Scholar 

  149. Li F, Tang H, Luo X, Li X, Luo K, Liu S, Liang J, Liao S, Zhong C, Zhan X, et al. Interaction gene set between osteoclasts and regulatory CD4(+) T cells can accurately predict the prognosis of patients with osteosarcoma. Cancer Sci. 2023;114:3014–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Zhang W, Dang K, Huai Y, Qian A. Osteoimmunology: The regulatory roles of T lymphocytes in osteoporosis. Front Endocrinol (Lausanne). 2020;11:465.

    Article  PubMed  Google Scholar 

  151. Sun W, Meednu N, Rosenberg A, Rangel-Moreno J, Wang V, Glanzman J, Owen T, Zhou X, Zhang H, Boyce BF, et al. B cells inhibit bone formation in rheumatoid arthritis by suppressing osteoblast differentiation. Nat Commun. 2018;9:5127.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Grcevic D, Sanjay A, Lorenzo J. Interactions of B-lymphocytes and bone cells in health and disease. Bone. 2023;168: 116296.

    Article  CAS  PubMed  Google Scholar 

  153. Fischer V, Haffner-Luntzer M. Interaction between bone and immune cells: Implications for postmenopausal osteoporosis. Semin Cell Dev Biol. 2022;123:14–21.

    Article  PubMed  Google Scholar 

  154. Walsh MC, Takegahara N, Kim H, Choi Y. Updating osteoimmunology: regulation of bone cells by innate and adaptive immunity. Nat Rev Rheumatol. 2018;14:146–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Duan L, Liang Y, Xu X, Xiao Y, Wang D. Recent progress on the role of miR-140 in cartilage matrix remodelling and its implications for osteoarthritis treatment. Arthritis Res Ther. 2020;22:194.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Lefebvre V, Dvir-Ginzberg M. SOX9 and the many facets of its regulation in the chondrocyte lineage. Connect Tissue Res. 2017;58:2–14.

    Article  CAS  PubMed  Google Scholar 

  157. Li C, Wang Q, Wang JF. Transforming growth factor-beta (TGF-beta) induces the expression of chondrogenesis-related genes through TGF-beta receptor II (TGFRII)-AKT-mTOR signaling in primary cultured mouse precartilaginous stem cells. Biochem Biophys Res Commun. 2014;450:646–51.

    Article  CAS  PubMed  Google Scholar 

  158. Pereira RC, Martinelli D, Cancedda R, Gentili C, Poggi A. Human articular chondrocytes regulate immune response by affecting directly T cell proliferation and indirectly inhibiting monocyte differentiation to professional antigen-presenting cells. Front Immunol. 2016;7:415.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Abe S, Nochi H, Ito H. Human articular chondrocytes induce interleukin-2 nonresponsiveness to allogeneic lymphocytes. Cartilage. 2017;8:300–6.

    Article  CAS  PubMed  Google Scholar 

  160. Wang YH, Zhao CZ, Wang RY, Du QX, Liu JY, Pan J. Correction: The crosstalk between macrophages and bone marrow mesenchymal stem cells in bone healing. Stem Cell Res Ther. 2022;13:524.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Xu H, Lee CW, Wang YF, Huang S, Shin LY, Wang YH, Wan Z, Zhu X, Yung PSH, Lee OK. The role of paracrine regulation of mesenchymal stem cells in the crosstalk with macrophages in musculoskeletal diseases: A systematic review. Front Bioeng Biotechnol. 2020;8: 587052.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Saldana L, Bensiamar F, Valles G, Mancebo FJ, Garcia-Rey E, Vilaboa N. Immunoregulatory potential of mesenchymal stem cells following activation by macrophage-derived soluble factors. Stem Cell Res Ther. 2019;10:58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Zhang Y, Bose T, Unger RE, Jansen JA, Kirkpatrick CJ, van den Beucken J. Macrophage type modulates osteogenic differentiation of adipose tissue MSCs. Cell Tissue Res. 2017;369:273–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Pajarinen J, Lin T, Gibon E, Kohno Y, Maruyama M, Nathan K, Lu L, Yao Z, Goodman SB. Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials. 2019;196:80–9.

    Article  CAS  PubMed  Google Scholar 

  165. Munoz J, Akhavan NS, Mullins AP, Arjmandi BH. Macrophage polarization and osteoporosis: A review. Nutrients. 2020;12:2999.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Loi F, Cordova LA, Pajarinen J, Lin TH, Yao Z, Goodman SB. Inflammation, fracture and bone repair. Bone. 2016;86:119–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Espagnolle N, Balguerie A, Arnaud E, Sensebe L, Varin A. CD54-Mediated interaction with pro-inflammatory macrophages increases the immunosuppressive function of human mesenchymal stromal cells. Stem Cell Reports. 2017;8:961–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Li Y, Kong N, Li Z, Tian R, Liu X, Liu G, Wang K, Yang P. Bone marrow macrophage M2 polarization and adipose-derived stem cells osteogenic differentiation synergistically promote rehabilitation of bone damage. J Cell Biochem. 2019;120:19891–901.

    Article  CAS  PubMed  Google Scholar 

  169. Khodayari S, Khodayari H, Amiri AZ, Eslami M, Farhud D, Hescheler J, Nayernia K. Inflammatory microenvironment of acute myocardial infarction prevents regeneration of heart with stem cells therapy. Cell Physiol Biochem. 2019;53:887–909.

    Article  CAS  PubMed  Google Scholar 

  170. Cho DI, Kim MR, Jeong HY, Jeong HC, Jeong MH, Yoon SH, Kim YS, Ahn Y. Mesenchymal stem cells reciprocally regulate the M1/M2 balance in mouse bone marrow-derived macrophages. Exp Mol Med. 2014;46: e70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Beird HC, Bielack SS, Flanagan AM, Gill J, Heymann D, Janeway KA, Livingston JA, Roberts RD, Strauss SJ, Gorlick R. Osteosarcoma. Nat Rev Dis Primers. 2022;8:77.

    Article  PubMed  Google Scholar 

  172. Zhao X, Wu Q, Gong X, Liu J, Ma Y. Osteosarcoma: a review of current and future therapeutic approaches. Biomed Eng Online. 2021;20:24.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Panez-Toro I, Munoz-Garcia J, Vargas-Franco JW, Renodon-Corniere A, Heymann MF, Lezot F, Heymann D. Advances in osteosarcoma. Curr Osteoporos Rep. 2023;21:330–43.

    Article  PubMed  PubMed Central  Google Scholar 

  174. O’Donoghue JC, Freeman FE. Make it STING: nanotechnological approaches for activating cGAS/STING as an immunomodulatory node in osteosarcoma. Front Immunol. 2024;15:1403538.

    Article  PubMed  PubMed Central  Google Scholar 

  175. He J, He Y, Biao R, Wei Y, Dong Z, Du J. STYK1 mediates NK cell anti-tumor response through regulating CCR2 and trafficking. J Transl Med. 2024;22:943.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Yu L, Zhang J, Li Y. Effects of microenvironment in osteosarcoma on chemoresistance and the promise of immunotherapy as an osteosarcoma therapeutic modality. Front Immunol. 2022;13: 871076.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Phillips D, Matusiak M, Gutierrez BR, Bhate SS, Barlow GL, Jiang S, Demeter J, Smythe KS, Pierce RH, Fling SP, et al. Immune cell topography predicts response to PD-1 blockade in cutaneous T cell lymphoma. Nat Commun. 2021;12:6726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Chen S, Lei J, Mou H, Zhang W, Jin L, Lu S, Yinwang E, Xue Y, Shao Z, Chen T, et al. Multiple influence of immune cells in the bone metastatic cancer microenvironment on tumors. Front Immunol. 2024;15:1335366.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Tomassetti C, Insinga G, Gimigliano F, Morrione A, Giordano A, Giurisato E. Insights into CSF-1R expression in the tumor microenvironment. Biomedicines. 2024;12:2381.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Chiabotto G, Grignani G, Todorovic M, Martin V, Centomo ML, Prola E, Giordano G, Merlini A, Miglio U, Berrino E, et al. Pazopanib and trametinib as a synergistic strategy against osteosarcoma: Preclinical activity and molecular insights. Cancers (Basel). 2020;12:1519.

    Article  CAS  PubMed  Google Scholar 

  181. Giordano G, Merlini A, Ferrero G, Mesiano G, Fiorino E, Brusco S, Centomo ML, Leuci V, D’Ambrosio L, Aglietta M, et al. EphA2 expression in bone sarcomas: bioinformatic analyses and preclinical characterization in patient-derived models of osteosarcoma Ewing’s Sarcoma and Chondrosarcoma. Cells. 2021;10:2893.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Schulte BC, Hwang C, Horvai AE, Olivier T. When destiny doesn’t pan out: Implications for the treatment of sarcomas after the agnostic approval of trastuzumab deruxtecan. Cancer. 2024;130:4020–4.

    Article  CAS  PubMed  Google Scholar 

  183. Limaiem F, Davis DD, Sticco KL. Chondrosarcoma. In: StatPearls. Treasure Island (FL): StatPearls; 2024.

    Google Scholar 

  184. Ingangi V, De Chiara A, Ferrara G, Gallo M, Catapano A, Fazioli F, Di Carluccio G, Peranzoni E, Marigo I, Carriero MV, Minopoli M. Emerging treatments targeting the tumor microenvironment for advanced chondrosarcoma. Cells. 2024;13:977.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Yin J, Ren P. New advances in the treatment of chondrosarcoma under the PD-1/PD-L1 pathway. J Cancer Res Ther. 2024;20:522–30.

    Article  PubMed  Google Scholar 

  186. Giordano G, Tucciarello C, Merlini A, Cutrupi S, Pignochino Y. Targeting the EphA2 pathway: could it be the way for bone sarcomas? Cell Commun Signal. 2024;22:433.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Xin B, Chen H, Zhu Z, Guan Q, Bai G, Yang C, Zou W, Gao X, Li L, Liu T. FBXO22 is a potential therapeutic target for recurrent chondrosarcoma. J Bone Oncol. 2024;46: 100605.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Strauss SJ, Berlanga P, McCabe MG. Emerging therapies in Ewing sarcoma. Curr Opin Oncol. 2024;36:297–304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Leavey PJ, Laack NN, Krailo MD, Buxton A, Randall RL, DuBois SG, Reed DR, Grier HE, Hawkins DS, Pawel B, et al. Phase III trial adding vincristine-topotecan-cyclophosphamide to the initial treatment of patients with nonmetastatic ewing sarcoma: A Children’s Oncology Group Report. J Clin Oncol. 2021;39:4029–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Heisey DAR, Jacob S, Lochmann TL, Kurupi R, Ghotra MS, Calbert ML, Shende M, Maves YK, Koblinski JE, Dozmorov MG, et al. Pharmaceutical interference of the EWS-FLI1-driven transcriptome by cotargeting H3K27ac and RNA polymerase activity in Ewing Sarcoma. Mol Cancer Ther. 2021;20:1868–79.

    Article  CAS  PubMed  Google Scholar 

  191. Chugh R, Ballman KV, Helman LJ, Patel S, Whelan JS, Widemann B, Lu Y, Hawkins DS, Mascarenhas L, Glod JW, et al. SARC025 arms 1 and 2: A phase 1 study of the poly(ADP-ribose) polymerase inhibitor niraparib with temozolomide or irinotecan in patients with advanced Ewing sarcoma. Cancer. 2021;127:1301–10.

    Article  CAS  PubMed  Google Scholar 

  192. Anderson P, Ghisoli M, Crompton BD, Klega KS, Wexler LH, Slotkin EK, Stanbery L, Manning L, Wallraven G, Manley M, et al. Pilot study of recurrent Ewing’s Sarcoma management with vigil/temozolomide/irinotecan and assessment of circulating tumor (ct) DNA. Clin Cancer Res. 2023;29:1689–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Subbiah V, Chawla SP, Conley AP, Wilky BA, Tolcher A, Lakhani NJ, Berz D, Andrianov V, Crago W, Holcomb M, et al. Preclinical characterization and phase I trial results of INBRX-109, a third-generation, recombinant, humanized, death receptor 5 agonist antibody, in chondrosarcoma. Clin Cancer Res. 2023;29:2988–3003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Luo W, Hoang H, Miller KE, Zhu H, Xu S, Mo X, Garfinkle EAR, Costello H, Wijeratne S, Chemnitz W, et al. Combinatorial macrophage induced innate immunotherapy against Ewing sarcoma: Turning “Two Keys” simultaneously. J Exp Clin Cancer Res. 2024;43:193.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Garcia-Monclus S, Lopez-Alemany R, Almacellas-Rabaiget O, Herrero-Martin D, Huertas-Martinez J, Lagares-Tena L, Alba-Pavon P, Hontecillas-Prieto L, Mora J, de Alava E, et al. EphA2 receptor is a key player in the metastatic onset of Ewing sarcoma. Int J Cancer. 2018;143:1188–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Sainz-Jaspeado M, Lagares-Tena L, Lasheras J, Navid F, Rodriguez-Galindo C, Mateo-Lozano S, Notario V, Sanjuan X, Garcia DMX, Fabra A, Tirado OM. Caveolin-1 modulates the ability of Ewing’s sarcoma to metastasize. Mol Cancer Res. 2010;8:1489–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Sainz-Jaspeado M, Huertas-Martinez J, Lagares-Tena L, Martin LJ, Mateo-Lozano S, de Alava E, de Torres C, Mora J, Del MXG, Tirado OM. EphA2-induced angiogenesis in ewing sarcoma cells works through bFGF production and is dependent on caveolin-1. PLoS ONE. 2013;8: e71449.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Basu MA, Chawla SP. Giant cell tumor of bone: An update. Curr Oncol Rep. 2021;23:51.

    Article  Google Scholar 

  199. Borkowska AM, Szumera-Cieckiewicz A, Szostakowski B, Pienkowski A, Rutkowski PL. Denosumab in giant cell tumor of bone: Multidisciplinary medical management based on pathophysiological mechanisms and real-world evidence. Cancers (Basel). 2022;14:2290.

    Article  CAS  PubMed  Google Scholar 

  200. Li H, Gao J, Gao Y, Lin N, Zheng M, Ye Z. Denosumab in giant cell tumor of bone: current status and pitfalls. Front Oncol. 2020;10: 580605.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Noguchi T, Sakamoto A, Murotani Y, Murata K, Hirata M, Yamada Y, Toguchida J, Matsuda S. Inhibition of RANKL expression in osteocyte-like differentiated tumor cells in giant cell tumor of bone after denosumab treatment. J Histochem Cytochem. 2023;71:131–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Lipplaa A, Dijkstra S, Gelderblom H. Challenges of denosumab in giant cell tumor of bone, and other giant cell-rich tumors of bone. Curr Opin Oncol. 2019;31:329–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Chen Z, Zhang C, Hong H, Xu W, Sha M, Ding Z. Potential alternative drug treatment for bone giant cell tumor. Front Cell Dev Biol. 2023;11:1193217.

    Article  PubMed  PubMed Central  Google Scholar 

  204. Toda Y, Kohashi K, Yamamoto H, Ishihara S, Ito Y, Susuki Y, Kawaguchi K, Kiyozawa D, Takamatsu D, Kinoshita I, et al. Tumor microenvironment in giant cell tumor of bone: evaluation of PD-L1 expression and SIRPalpha infiltration after denosumab treatment. Sci Rep. 2021;11:14821.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Fan M, Zhang G, Xie M, Liu X, Zhang Q, Wang L. Siglec-15 as a new perspective therapy target in human giant cell tumor of bone. Curr Oncol. 2022;29:7655–71.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Chatterjee C, Schertl P, Frommer M, Ludwig-Husemann A, Mohra A, Dilger N, Naolou T, Meermeyer S, Bergmann TC, Alonso CA, Lee-Thedieck C. Rebuilding the hematopoietic stem cell niche: Recent developments and future prospects. Acta Biomater. 2021;132:129–48.

    Article  CAS  PubMed  Google Scholar 

  207. Bello AB, Park H, Lee SH. Current approaches in biomaterial-based hematopoietic stem cell niches. Acta Biomater. 2018;72:1–15.

    Article  CAS  PubMed  Google Scholar 

  208. Severn CE, Eissa AM, Langford CR, Parker A, Walker M, Dobbe JGG, Streekstra GJ, Cameron NR, Toye AM. Ex vivo culture of adult CD34(+) stem cells using functional highly porous polymer scaffolds to establish biomimicry of the bone marrow niche. Biomaterials. 2019;225: 119533.

    Article  CAS  PubMed  Google Scholar 

  209. Oliveira CS, Carreira M, Correia CR, Mano JF. The therapeutic potential of hematopoietic stem cells in bone regeneration. Tissue Eng Part B Rev. 2022;28:379–92.

    Article  CAS  PubMed  Google Scholar 

  210. Pietrzyk-Nivau A, Poirault-Chassac S, Gandrille S, Derkaoui SM, Kauskot A, Letourneur D, Le Visage C, Baruch D. Three-dimensional environment sustains hematopoietic stem cell differentiation into platelet-producing megakaryocytes. PLoS ONE. 2015;10: e0136652.

    Article  PubMed  PubMed Central  Google Scholar 

  211. Pereira AL, Galli S, Nombela-Arrieta C. Bone marrow niches for hematopoietic stem cells. Hemasphere. 2024;8: e133.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This study was supported by the National Natural Science Foundation of China (81972085, 82172465), China University Industry-University-Research Innovation Fund (2021JH037), the Natural Science Foundation of Guangdong Province (2021A1515010706, 2023A1515010102), Guangdong Provincial Key Clinical Discipline-Orthopedics (2000005), the Sanming Project of Shenzhen Health and Family Planning Commission (SZSM202311008), Shenzhen Science and Technology Planning (GJHZ20210705142007023), the Shenzhen Key Medical Discipline Construction Fund (SZXK025), Scientific Research Foundation of Shenzhen Health Economics Association (2023100, 2023101, 2023106). Team-based Medical science Research Program (2024YZZ13).

Author information

Authors and Affiliations

Authors

Contributions

Zeqi Huang, Zoya Iqbal, Zhe Zhao, Jianquan Liu, Maryam Shabbir, Ayesha Mahmood, Yujie Liang, Wencui Li and Zhiqin Deng participated in the literature review, Zeqi Huang and Yujie Liang performed the figure design, and Zeqi Huang and Zoya Iqbal wrote the manuscript. The authors read and approved the final manuscript.

Corresponding authors

Correspondence to Yujie Liang, Wencui Li or Zhiqin Deng.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, Z., Iqbal, Z., Zhao, Z. et al. Cellular crosstalk in the bone marrow niche. J Transl Med 22, 1096 (2024). https://doi.org/10.1186/s12967-024-05900-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05900-6

Keywords