Skip to main content

The mechanisms of mutual relationship between malignant hematologic cells and mesenchymal stem cells: Does it contradict the nursing role of mesenchymal stem cells?

Abstract

Mesenchymal stem/stromal cells (MSCs) are known as the issue in biology because of some unpredictable characteristics in the different microenvironments especially in their bone marrow niche. MSCs are used in the regenerative medicine because of their unique potentials for trans-differentiation, immunomodulation, and paracrine capacity. But, their pathogenic and pro-survival effects in tumors/cancers including hematologic malignancies are indisputable. MSCs and/or their derivatives might be involved in tumor growth, metastasis and drug resistance in the leukemias. One of important relationship is MSCs and hematologic malignancy-derived cells which affects markedly the outcome of disease. The communication between these two cells may be contact-dependent and/or contact-independent. In this review, we studied the crosstalk between MSCs and malignant hematologic cells which results the final feedback either the progression or suppression of blood cell malignancy.

Graphical abstract

Video abstract

Background

Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are a rare population of non-hematopoietic multipotent cells that are recognizable and sortable by being positive for MSC markers such as CD90, CD73 and CD105 and being negative for hematopoietic markers like CD34 and CD45 [1, 2]. It has been revealed that it is not just the physical support provided by MSCs in microenvironment for target cells, but a bidirectional conversation between them is routed [3, 4]. A substantial number of studies declare that BM-MSCs adopt several methods to establish this communication. These mechanisms can be divided into two main categories: contact-dependent including tunneling nanotubes (TNTs) [5, 6], gap junctions [7], and contact-independent including paracrine activity and microvesicle/exosomes delivery through which microRNA (miRNA), immuno-modulatory molecules and mitochondria can be transferred [8,9,10,11,12]. MSCs are mainly being used in the regenerative medicine [13,14,15]. But in terms of malignancy, it is a subject of controversial that whether MSCs are pro- or anti-tumorigenic [16]. However, to digress from malignancies, MSCs are also used in myocardial infarction (MI) [17]. They play an indirect role in MI by recruitment of macrophages in angiogenesis to promote tissue regeneration [18]. The study of these effects and conversations provides a fertile ground of investigation [19].

Returning to malignancies, although MSCs have shown therapeutic properties mainly by their potentials for trans-differentiation, immunomodulation, and apoptosis induction [2, 20, 21], their pathogenic, leukemogenic and pro-survival effects in hematologic malignancies are indisputable [22, 23]. Cellular mechanisms include RNA processing, ubiquitin–proteasome pathway, cell cycle regulation, cellular stress and non-canonical Wnt signaling are modulated in the leukemia cell lines co-cultured with MSCs [24].

Indeed, MSCs have been suspected of being the culprit of elevated tumor growth, metastasis and drug resistance in the leukemias [25]. Particularly in chronic lymphocytic leukemia (CLL) cells, survival and drug resistance signals as well as CLL-cell trafficking and tissue homing signals are under bone marrow stromal cells (BMSCs) control [26]. Even in a mouse model of the pre-leukemic disorder Schwachman-Diamond syndrome (SDS), genotoxic stress and subsequently DNA damage response (DDR) activation in hematopoietic stem and progenitor cells have been reported as the main consequence of abnormal activity of mesenchymal cells [27]. Therefore, this study covers a lot of ground in this matter and offers a full explanation of mechanisms by which MSCs counter treatment strategies. Herein, the contact-dependent and contact-independent mechanisms that involve in the MSC and leukemic cells conversation are presented.

Contact-dependent

Tunneling nanotubes (TNTs)

TNTs are intercellular transient structures of about 50–200 nm in diameter which are made of polymerization of F-actin; thereby, they are naturally vanishing and henceforth uncontrollable. Likewise, biomolecules and cellular organelles can be transported through them [28].

First of all, chemoresistance can be modulated in other solid tumor cells like SKOV3 ovarian cancer cells and MCF7 breast cancer cells as a consequence of preferential transfer of mitochondria from endothelial to cancer cells through TNTs modulates [29]. Pinto and et al. have reviewed that TNT-like connections are used by cancers to modify their potential chemoresistance, migration, metabolism, metastasis and angiogenesis [30].

However, in the leukemias TNTs serve the reliable infrastructure for trafficking both vesicle and protein to leukemic cells [31]. This type of intercellular relationship is forged, and as it has been proven in B cell acute lymphoblastic leukemia (B-ALL), it is not a monologue delivered by MSCs but dialogues can be held. As an example, using TNTs, BM-MSC secretome profile is pervasively converted to a leukemia pro-survival profile in B-lymphoblastic leukemia [32]. Bidirectional cytoplasmic transport has also been observed between MSCs and activated benign T cells which exerts suppressive effect on the proliferation rate and IFN-γ production of T cells [33].

Furthermore, through a TNT routed communication, primary B-cell precursor ALL (BCP-ALL) cells deliver autophagosomes, mitochondria, intercellular adhesion molecule 1 (ICAM1) and other lipophiles to MSCs leading to cytokine secretion, leukemic cell survival and drug resistance [34]. These pro-survival cytokines include interferon-γ-inducible protein 10 (IP-10), CXCL10, IL-8, monocyte chemotactic protein-1 (MCP-1) and CCL2 [35]. Disruption of TNTs would physically affect this support and significantly re-sensitizes BCP-ALL cells to chemotherapeutics like prednisolone [35]. However, being exposed to chemotherapeutic drugs, mitochondrial transfer from Jurkat cells, a T cell acute lymphoblastic leukemia (T-ALL) cell line, to MSCs is promoted. Although, it is primarily conducted by contact-dependent manners including TNTs and gap junctions (GJs), nevertheless microvesicles (contact-independent) are also to some extent responsible for this trafficking [36]. In fact, Jurkat cells exhibit a trafficking arabinoside (ARA-C) or methotrexate. This leads to a substantial promotion of Jurkat cells' survival, due to a dramatic reduction of reactive oxygen species (ROS) levels inside them [36, 37]. The resulting increased production of mitochondrial adenosine triphosphate (ATP), mitochondrial transfer leads to increased chemoresistance [38].

By the same token, in acute myelogenous leukemia (AML), mitochondrial transfer primarily through TNT has been contingently associated with chemoresistance [39]. Both leukemic blasts and leukemia initiating cells of AML are given the privilege of further survival in battle with some chemotherapies by the TNT-routed mitochondrial transfer from MSC [40]. By up to 14%, mitochondrial mass in AML cells increases in co-cultures with MSCs. Consequently, cytotoxic effects of the nucleoside analog ARA-C would be hedged against AML cell lines [40].

To enhance transferring from MSC to AML blasts through TNT, mitochondrial biogenesis in MSC is stimulated by AML-derived NADPH oxidase 2 (NOX2) superoxide. Correspondingly, this facilitation can be taken place in multiple myeloma (MM) cells by CD38 expressing MM cells [41]. Inhibition of NOX2 is characterized by a downward trend in mitochondrial transfer and an upward trend in AML cells apoptosis [42]. Autophagy, on the other hand is increased in AML cells chiefly because autophagosomes transfer from MSCs to AML cells using TNTs [32].

Tyrosine kinase inhibitors and interferon‐α used in chronic myeloid leukemia (CML) cell lines increase TNT formation and cell adhesion [43]. The so-called stroma-mediated imatinib resistance would be resulted from this TNT formation. To clarify, CML cells receive cellular vesicles from and send them out to MSCs through TNTs. These vesicles finally attenuate imatinib mediated caspase activity and thereby apoptosis [44]. Notably in CML, TNTs facilitate exosomes transportation and exosomes in turn stimulate TNT formation; so, they can synergistically interact and multiply their communication [45].

Turning to lymphoma, we must note that malignant B cells upregulate BCL-2 family proteins following receiving soluble factors from MSCs. Activation of the oncogenic pathways like NOTCH1 signaling is also stimulated by MSCs reported in different B cell malignancies, MM and CLL [46, 47]. Examples of MSCs and malignant hematologic cells communications via TNTs have been summarized in Table 1

Table 1 Communication between MSCs and hematologic malignancies/cell lines through tunneling nanotubes (TNTs) and its effects

Gap junctions

Gap junctions (GJs) are comprised of two hemichannels (HCs) or connexons which in turn composed of arrays of connexin proteins. Ions, small metabolites, and organelles can be transferred through GJs. GJ intercellular communication (GJIC) in the HSC niche may lead to improvement of cellular bioenergetics, and rejuvenates the damaged recipient cells [50].

In normal hematopoiesis, mitochondria can transfer from BMSC to HSC using GJ by which GJs modulate granulopoiesis and differentiation to myeloid blood cell precursor. Hematopoietic stem/progenitor cells (HSPCs) quiescence and stemness are also determinable by GJ communication named as connexin-32 (Cx32). HSC quiescence retention and survival in the BM is dependent on CXCL12 secretion which is regulable by the expression of both connexin-43 (Cx43( and connexin-45 (Cx45) in MSC. During stress, Cx43 transfers ROS to BMSC which reduces HSC senescence [50].

Turning to malignancy, in co-culture with MM cells, MSCs exhibit an abrupt increase in Cx43 level representative of improved GJ-mediated intercellular communication [51]. Cx43 has also been postulated to be a putative player of adhesion and migration of MM cells demonstrated in primary MM cells and cell lines RPMI 8226, U266, and XG-7 which can finally increase cell proliferation and chemoresistance [18]. Adhesion and migration of MM cells is blocked by the gap junction blocker 18α-glycyrrhetinic acid (18α-GA) which decrease stromal cell-derived factor-1α (SDF-1α) secretion [51]. Divagating from MM, HL-60 and PBL-985 cells would see a downward trend in differentiation potential when GJ communication has not been abrogated [50, 52].

Carbenoxolone-induced GJ disruption could interfere with MSC and different malignant hematologic cell line communication and alters drug resistance pattern [53]. It indicates the important role of GJ between these cells in the outcome of leukemia treatment.

Cx26, Cx32, Cx37, Cx43, and Cx45 are responsible for exponentially elevated chemoresistance and substantially reduced apoptosis in primary AML cells. Leukemia pathogenesis is interconnected with connexin-based modifications on target cells. For example, mitochondria can transfer in a Cx43-mediated manner which affect adversely not only the pathogenesis but also chemoresistance [50]. Therefore, MSCs induced chemoresistance can be modulated by disruption of gap junctions in AML [53].

A significant amounts of prostaglandin E2 (PGE2), which suppresses DNA damage-induced p53 accumulation, is released from the HCs of stromal cells leading to promoted survival and metastasis of cancer cells [54]. However, having the permeability to small molecules and macromolecules, Cx43 may provide a target for cytoplasmic drug delivery [55]. Table 2 represents details of the GJ-mediated crosstalk between MSCs and malignant hematologic cells that has reported in different studies.

Table 2 Crosstalk between MSCs and hematologic malignancies/cell lines via gap junction and its effects

Contact-independent

Paracrine activity

MSC produce cytokines such as IL-6, IL-11, SCF, TPO, Flt-3 ligand, CXCL12, G-CSF, GM-CSF and M-CSF [58,59,60,61] to support hematopoiesis. On the other hand, HSC quiescence is affected by Wnt released from MSC. Wnt expression in HSC also downregulated kit ligand, angiopoietin-1, CXCL12 and vascular cell adhesion molecule 1 (VCAM-1) [62, 63].

To turn to malignant hematopoiesis, BM-MSCs mainly produce Wnt ligands which leads to the intracellular accumulation of β-catenin. Gene expression of several downstream growth factors are subsequently elevated and proliferation of leukemia stem cells (LSCs) is guaranteed by this way [64, 65]. To elucidate, the effects of Wnt/β-catenin signaling as a pro-growth signal is essentially counteracted by the bone morphogenetic protein (BMP) anti-growth signals. Imbalance toward higher growth rate results in a leukemogenic phenotype [66]. However, dealing with Wnt/β‐catenin signaling, scientists have found this pathway to be complex and regulated by MSCs themselves. Interferon‐β (IFN‐β) released from MSCs exhibits anti‐tumorigenic effects in erythroleukemic cells based on its ability to negatively regulate Wnt/β‐catenin signaling pathways [67]. Dickkopf‐1 (DKK‐1) is also a negative regulator of Wnt signaling pathway and has antiproliferative activity in MM [68]. Considering the stimulatory effect of MSCs in production of DKK-1, IL-6, and IL-10, a potential role has been ascribed to the crosstalk between myeloma and MSCs in the development of disease into a bone lytic phase [57, 69]. Interestingly, in co-culture studies with multiple myeloma-derived mesenchymal stem cells (MM-MSCs), granulocytic-myeloid-derived suppressor cells (G-MDSCs) have been examined. MM-MSC educated G-MDSCs demonstrate supportive effects in MM by upregulation of immune-suppressive and proangiogenic factors including arginase 1 (ARG1), tumor necrosis factor α (TNF-α), and prokineticin 2 (PROK2) [70]. Besides, MSC ensures MM cell survival, disease progression, and drug resistance having upregulated levels of gene expression of angiogenic and growth factors such as CD40/40L, VCAM-1, ICAM-1, lymphocyte function-associated antigen-3 (LFA-3), and immunomodulated level of cytokines: increased IL-6 and reduced IL-10 [71]. Pro-angiogenic profile accompanied by anti-osteogenic pattern in MM cells co-cultivated by MM-MSCs can be a consequence of increased the vascular endothelial growth factor (VEGF) and IL-6 expression. As a matter of interest, this phenomenon is followed from activation of Notch signaling in MM-MSCs [72].

Regarding other cytokines produced by MSCs, they secrete promyelocytic leukemia protein followed by production of pro-inflammatory molecules, including CXCL1 and IL-6 which is considered as the major cause of leukemogenesis in the different types of leukemia [73]. For the most part, MSC secreted factors, especially IL-6, shelter CML cells from imatinib-induced apoptosis basically through NFκB-mediated signaling [74]. Chemoresistance in the diffuse large B-cell lymphoma (DLBCL) can be acquired by MSC secretion of IL-6 and upregulation of IL-17A [75].

In CLL, MSCs also demonstrate protective activities against cytotoxic effects of Forodesine [76]. To illustrate, interaction of MSC with CLL cells increases the production platelet-derived growth factor (PDGF), which binds to its receptor, PDGFR, leading to secretion of VEGF and making an angiogenic switch, associated with drug protection and disease progression [77]. Compared to MDS-derived mesenchymal stromal cells (MDS-MSCs), MSCs from B-CLL patients produce aberrant SDF-1, B-cell activating factor (BAFF), and transforming growth factor β (TGF-β) resulting in exponentially promoted normal B-cell proliferation and IgG production [78]. Elevated VEGF and hypoxia-inducible factor 1 (HIF-1) production is representative for proangiogenic profile and therefore additional CLL cell survival and resistance to rituximab/alemtuzumab [71].

Disease progression resulted from shifting to proangiogenic profile is the fatal outcome of interaction between conditioned medium (CM) obtained from CLL cells (CLL-CM) and MSCs. PDGFR in MSCs is converted to the active form after exposure to CLL-CM. Microenvironment must face devastating consequences of this phenomenon including MSC proliferation and MSC VEGF production [79]. Finally, survival of CLL cells is also insured by the interaction between their hepatocyte growth factor receptor (c-MET) and hepatocyte growth factor secreted by MSCs [80]. Some important MSCs-derived molecules and their related effects on HSCs and leukemic cells were shown in Table 3.

Table 3 Paracrine effects of MSCs on HSCs and different leukemic cells

Chemokines and bio active molecules

There are also some chemokines that are regulated by MSCs. First of all, through CXCL12-CXCR4 interaction between MSC and CML cells (respectively), imatinib-induced cell death is reduced as a consequence of attenuated caspase-3 activity [76]. Niches with CXCL12 devoid of MSCs, cannot support the LSCs from tyrosine kinase inhibitor (TKI) treatment, while CXCL12 + MSC niches offer a full guarantee for LSCs to maintain quiescent and TKI-resistant [83].

Secondly, CXCL8 derived from MSCs supports the survival and proliferation of AML cells through the PI3K/AKT pathway [84]. By the same token, via activation of NF-κB, MSC is involved in the residual disease maintenance in AML and on the other hand in therapy-resistance. This activation of NF-κB may be the consequence of interaction of VCAM-1 on MSC and its ligand, VLA-4, on leukemic cells [85]. Indeed, the underlying molecular mechanisms in BM niche by which the drug resistance and disease relapse are caused in AML include SDF-1/CXCL12, Wnt/β-catenin, VCAM/VLA-4/NF-κB, CD44, and hypoxia [86]. Axl is a member of the Tyro3 and has been approved of prognostic value and therapeutic target in AML that has been claimed as a mediator in the paracrine signaling between the leukemia cells and BM-MSCs. The expression of Axl ligand, growth arrest–specific gene 6 (Gas6), on MSCs can be elicited by AML cells.[87].

Other factors including Periostin (POSTN) is a multifunctional extracellular component. BM-MSC-derived POSTN promotes B-ALL cell-derived CCL2 which increases the leukemia burden [88]. Lumican (LUM) is an extracellular matrix protein secreted by MSCs. LSCs such as Nalm-6 (an ALL cell line) acquires anti-apoptotic properties and resistance to chemotherapy by downregulation of LUM expression in BM-MSCs [89].

Bone destruction in MM is mainly orchestrated by osteoclasts that undergo differentiation induced by the production of CCL3 and CCL4, matrix metalloproteinases (MMP)-13, IL-1, IL-3, IL-6 and IL-17 released by MSCs [72]. Conversely, AML cells shift the niche towards an osteoblastic one by the induction of connective tissue growth factor (CTGF) expression in BM-MSCs [90].

Co-cultured with MSCs, CML cells reduce caspase-3 activation and modulate Bcl-XL (anti-apoptotic protein) expression after treatment with imatinib which signify MSC-mediated protection of CML cells [91, 92]. This has been proven to be interceded with CXCR4/CXCL12; hence, combinational therapy with anti-CXCR4 antagonists and TKIs may represent a powerful approach in the treatment of CML [93].

Apart from intracellular signaling pathways, inhibiting the intercellular trafficking routes provides a promising therapeutic approach in leukemia. Using AMD3100 for example, the SDF-1α/CXCR4 axis is interrupted leading to hindrance of intercellular trafficking of CLL cells, and disturbance of microenvironment-mediated support [94]. AMD3100 is the first generation CXCR4 antagonist; therefore, it can inhibit proliferation of HSC and trafficking of leukocytes. However, BL8040 is the CXCR4 new generation inhibitor exhibiting higher affinity than AMD3100. As it has been presented in Table 4, chemokines and biomolecules interfere with malignant hematologic cells using different mechanisms.

Table 4 MSCs-secreted chemokines/biomolecules and their impacts on the hematologic malignancies

Microvesicles and exosomes

Microvesicles (MVs) and exosomes shed from MSCs membrane [10] and affect on different cell processes. Cell viability, clonogenic capacity and miRNA and gene expression profile of CD34+ cells in patients with MDS were all modified after receiving MVs derived from MSCs [49].

On the other hand, BM-MSCs ability to support CD34+ cells declines, after getting affected by extracellular vesicles (EVs) containing miR-7977 derived from AML/MDS CD34+ cells. miR-150 EVs target the CXCR4/SDF-1 axis which is fundamental for retention and differentiation of HSPC in BM. Instructed by human primary MDS cells, normal donor MSCs (ND-MSCs) adopt MDS-desirable features such as high expression of proangiogenic factors (VEGFA, IGFs, and EGFs) and mediators of fibrosis (LOXL, TGF-β, and LIF) [49].

Exosomes can be defined as the small, extracellular vesicles carrying a variety of biologic molecules, including proteins, DNA, mRNA and non-coding RNA. These proteins include antigen presenting molecules, adhesion molecules, membrane transport and fusion molecules, cytoskeletal proteins, pyruvate kinase, histones and others [10].

For the first consideration, MSC-derived EVs in the kidney, neurological, cardiovascular and liver diseases are of precious value that influence disease trajectory, patient survival and treatment strategy [96]. Secondly, cell-fate determination in stem cells is an EV-mastered process [97]. Modified by EVs, cancer stem cells (CSCs) and normal HSCs can develop and differentiate to various hematologic malignancies. These EVs are secreted by MSCs reprogrammed by CSCs and the neoplastic cells [98]. EVs content also modifies CD34+ cells viability as well as colony forming unit-granulocyte monocyte (CFU-GM) production. Precisely, some microRNAs like miR-10a and miR-15a are overexpressed in EVs from MSCs of MDSs patients and transferred to CD34+ cells. Modifying the expression of MDM2 and P53 genes, these microRNAs augment cell viability and increase clonogenic capacity [99]. On Immune cells, they induce immunosuppression [100]. EVs from MSC promote both proliferation and apoptosis of regulatory T cells [101]. They decrease Th17 cells and increase regulatory T cells on the peripheral blood mononuclear cells [102].

Based on studies on K562 cells-derived exosomes, these EVs may directly stimulate the target cells or transfer receptors between cells. They may deliver functional proteins and transfer the genetic materials like mRNA, miRNA, or transcription factors to target cell [103]. Another CML cell line, LAMA84, generates EVs that have effects on the human vascular endothelial cells leading to ICAM-1, VCAM‐1, and IL‐8 expression upregulation which indeed shift the tumor microenvironment (TME) to a pro-angiogenic pattern and therefore unfavorable prognosis [104].

Enhancement of angiogenesis is mainly mediated by the well-known pro-angiogenic factors such as VEGF, basic fibroblast growth factor (bFGF), and angiopoietin-1 secreted by the MM cells or stromal cells interacting with MM cells. Osteoclast differentiation and osteoclast bone resorption activity in MM is modulated and supported by MM cell-derived exosomes containing osteoclast activating factors which in turn enhance MM cell growth and survival by secretion of IL-6 and B-cell-activating factor [105].

Generally speaking, escaping from spontaneous or drug-induced apoptosis, migrating in higher rate and modifying genes more suitably are the main results of transferring EVs from leukemia patient MSCs compared to EVs from healthy donor MSCs [106]. IL-6 and IL-8 inhibit hematopoiesis by downregulating the CXCL12, angiopoietin 1, and kit ligand. In hematologic malignancies, IL-6 and IL-8 are upregulated in MSCs and are delivered to microenvironment by EVs [71].

LAMA84-derived exosomes promote IL-8 secretion in the MSC cell line, HS5, leading to enhance the survival, proliferation, and migration of LAMA84 cells in vitro. [71]. Based on a study about K562 cell, K562 exosomal miR-711 has been credited for suppressed adhesion abilities of BM-MSCs because of the fact that miR-711 is capable of silencing CD44—an adhesion molecule‒expression in BM-MSCs [107].

Exosomes from BM-MSCs contain miR-222-3p which is responsible for interferon regulatory factor 2/inositol polyphosphate 4-phosphatase type II (IRF2/INPP4B) signaling inhibition and has been greatly observed in co-culture with AML cell line [108]. IRF2/INPP4B signaling is involved in autophagy and apoptosis [109]. Exosomes secreted by AML cells alter the behavior of MSCs [110]. AML cell resistance to TKIs is effectively guaranteed by TGF-β1, miR-155, and miR-375 rich exosomes released by BM-MSC. Similarly, exosomes rich in miR-150 can disrupt the CXCR4/CXCL12 axis; disruption of CXCR4/CXCL12 axis supports the leukemia growth. These EVs are derived from AML cells and destined to be taken up by BM-MSCs [111].

Another in vitro study clarify that exosomes from AML cell lines HEL 92.1.7, HL-60, MOLM-14, and U937 transfer mRNA of insulin-like growth factor 1 receptor (IGF1R), matrix metalloproteinase 9 (MMP-9), nuclear matrix protein 1 (NPM1), CXCR4, and internal tandem duplication mutations in FLT3 (FLT3-ITD) into BM-MSC [71].

By the same token, exosomes derived from MM-MSC aim to ensure disease progression in vivo by delivery of IL-6, CCL2, and fibronectin and by attenuating the expression of the tumor suppressor miR-15a [71].

Tax viral oncoprotein of human T-cell lymphotropic virus type I (HTLV-I) causes adult T-cell leukemia/lymphoma (ATL). BM-MSCs pick exosomes up from ATL cells containing the Tax oncoprotein and leading to reduced MSC stemness and improved angiogenesis due to the multiplied levels of VEGF, CXCR4, and MMP-9 [71]. IL-8 secretion is initiated by CML cell-derived exosomes and finally contributes to CML cell survival [112].

Mechanistically, it has been proposed that the content of tumor-suppressor miR-15a in MSC-EVs is determinative of the MSC communication with MM cells. Decreased miR-15a content in MM-MSCs induces tumor growth and promotes myeloma dissemination [113, 114]. Proliferation, cancer-associated fibroblast (CAF) transformation, and IL-6 secretion of MSCs increases in co-culture with MM cells and these have been partially guided by miR-21 and miR-146a delivered by MM cells [115]. Minimal residual disease (MRD) after treatment can be monitored by measuring the circulating EVs in MM. Remarkably, transforming from monoclonal gammopathy of undetermined significance into symptomatic myeloma can gain the advantage of predictability by identifying and measuring the circulating EVs [116].

Based on both in vitro and in vivo studies, the leukemia-surviving subpopulation of MSCs in CLL cells is created and developed following secretion of protein- and miRNA-containing exosomes by CLL cells [32]. MSCs from CLL patients support in vitro neoplastic B cell survival [117]

EVs would be brought highly on agenda considering the fact that tumor stage, risk of recurrence, drug resistance, and overall clinical outcome of patients correlate to a great extent with number, phenotype and the molecular content of EVs [116]. Table 5 indicates some of studies and their reports about MV-mediated communication of MSCs and malignant cells.

Table 5 Interaction between MSCs and hematologic neoplasms by microvesicles transferring

Discussion

MSCs could communicate with malignant hematologic cells by different contact-dependent and/or contact-independent mechanisms. Notably in CML, TNTs facilitate exosomes transportation and exosomes in turn stimulate TNT formation; so, they can synergistically interact and multiply their communication [45]. However, having permeability to small molecules and macromolecules, Cx43 may provide a target for the cytoplasmic drug delivery [55]. Inhibition of oxidative phosphorylation (Oxphos) pathway in mitochondria also contributes to drug-resistance of AML based on the fact that TNT formation and mitochondrial transfer from BM-MSCs to AML is facilitated and promoted in this way [118]. In Jurkat cells, MSC-induced chemoresistance can be controlled by inhibition of mitochondrial transfer [36]. On the other hand, TNT formation is downregulated by NF-κB inhibitor BAY-117082 in AML [119]. Currently, BM-MSCs are found to be able to significantly enhance the drug resistance to various chemotherapy drugs, such as vincristine and cytarabine in ALL cells [120].

To clarify, in TME, MSCs-derived MVs can block the anti-tumor activity on immune cells and/or converts them into suppressor cells [111]. Another ascribed anti-tumor activity to MSCs is restoration of BM microenvironment via reprogramming the host macrophages [121]. Furthermore, MSC can inhibit the responses to alloreactive T lymphocytes as well as proliferation and cytotoxicity of natural killer (NK) cells [122].

Surprisingly, MSCs play an essential role for leukemia progression and chemoresistance by mitochondrial transfer, though the fate of transferred mitochondria in leukemic cells remains unclear. MSCs from patients with MDS and AML have a wide range of chromosomal aberrations, genetic and transcriptomic alterations. Deficiency of focal adhesion kinase (FAK) in MDS-MSCs correlates with ineffective hematopoiesis as it regulates the adhesion and mobility of cells [123].

It has been by the way proposed that the quiescence of AML blasts is ensured and outlasted in coculture with MSC resulting in increased leukemic survival in the presence of cytarabine [124]. Primary human AML cells remain proliferative for long-term by growth-enhancing effects of normal MSCs which is mediated by increased phosphorylation of the mammalian or mechanistic target of rapamycin (mTOR) and its downstream targets [125]. Diminished apoptosis is representative of tumor promoting effects of MSCs on MM cells and generally results from downregulation in caspase‐3 and poly (ADP‐ribose) polymerase expression which is associated with and mediated by enhanced AKT and ERK activities in MM cells [126].

Conclusions

In summary, bidirectional relationship between MSCs and hematologic malignancy-derived cells has different contact-dependent and contact-independent mechanisms. These cross-talks affect disease progression and outcome. The fate of malignant cells, drug resistance conditions, MRD status and other cellular processes are regulated by the MSC behavior. There are many studies conducted to understand the exact underlying mechanisms of MSCs and malignant hematologic cells communication. Their results could be applicable to design an improved treatment protocol and ameliorated patient’s survival. Hence, focus on this field and conducting additional studies or review with more confirmed information are emphatically suggested in this regard. We finally can infer that MSC does not behave similarly against different malignant hematologic cells and it basically extracted from the diverse responses and signals emitted from MSC in TME. It seems that the nursing role of MSCs in one hematologic neoplasm may be reversed in another by tumor progression and anti-apoptotic benefit.

Availability of data and materials

Not applicable.

Code availability

Not applicable.

Abbreviations

MSCs:

Mesenchymal stem/stromal cells

BM-MSCs:

Bone marrow-derived mesenchymal stromal cells

TNTs:

Tunneling nanotubes

miR:

MicroRNA

MI:

Myocardial infarction

CLL:

Chronic lymphocytic leukemia

BMSCs:

Bone marrow stromal cells

SDS:

Schwachman–Diamond syndrome

DDR:

DNA damage response

B-ALL:

B cell acute lymphoblastic leukemia

BCP-ALL:

B-cell precursor ALL

ICAM1:

Intercellular adhesion molecule 1

IP-10:

Interferon-γ-inducible protein 10

MCP-1:

Monocyte chemotactic protein-1

T-ALL:

T cell acute lymphoblastic leukemia

GJs:

Gap junctions

ARA-C:

Cytosine arabinoside

ROS:

Reactive oxygen species

AML:

Acute myelogenous leukemia

NOX2:

NADPH oxidase 2

MM:

Multiple myeloma

CML:

Chronic myeloid leukemia

HCs:

Hemichannels

GJIC:

GJ intercellular communication

HSPCs:

Hematopoietic stem/progenitor cells

Cx:

Connexin

SDF-1α:

Stromal cell-derived factor-1α

PGE2 :

Prostaglandin E2

VCAM-1:

Vascular cell adhesion molecule 1

BMP:

Bone morphogenetic protein

LSCs:

Leukemia stem cells

DKK‐1:

Dickkopf‐1

MM-MSCs:

Multiple myeloma-derived mesenchymal stem cells

G-MDSCs:

Granulocytic-myeloid-derived suppressor cells

ARG1:

Arginase 1

TNF-α:

Tumor necrosis factor α

PROK2:

Prokineticin 2

LFA-3:

Lymphocyte function-associated antigen-3

VEGF:

Vascular endothelial growth factor

DLBCL:

Diffuse large B-cell lymphoma

PDGF:

Platelet-derived growth factor

MDS-MSCs:

MDS-derived mesenchymal stromal cells

BAFF:

B-cell activating factor

TGF-β:

Transforming growth factor β

HIF-1:

Hypoxia-inducible factor 1

CM:

Conditioned medium

TKI:

Tyrosine kinase inhibitor

Gas6:

Growth arrest–specific gene 6

POSTN:

Periostin

LUM:

Lumican

MMP:

Matrix metalloproteinase

EVs:

Extracellular vesicles

ND-MSCs:

Normal donor MSCs

CSCs:

Cancer stem cells

CFU-GM:

Colony forming unit-granulocyte monocyte

TME:

Tumor microenvironment

bFGF:

Basic fibroblast growth factor

IRF2/INPP4B:

Interferon regulatory factor 2/inositol polyphosphate 4-phosphatase type II

IGF1R:

Insulin-like growth factor 1 receptor

FLT3-ITD:

Internal tandem duplication mutations in FLT3

HTLV-I:

Human T-cell lymphotropic virus type I

ATL:

Adult T-cell leukemia/lymphoma

CAF:

Cancer-associated fibroblast

MRD:

Minimal residual disease

FAK:

Focal adhesion kinase

Oxphos:

Oxidative phosphorylation

mTOR:

Mammalian (mechanistic) target of rapamycin

References

  1. Crippa S, Santi L, Bosotti R, Porro G, Bernardo ME. Bone marrow-derived mesenchymal stromal cells: a novel target to optimize hematopoietic stem cell transplantation protocols in hematological malignancies and rare genetic disorders. J Clin Med. 2020;9(1):2.

    Article  CAS  Google Scholar 

  2. Amiri F, Jahanian-Najafabadi A, Roudkenar MH. In vitro augmentation of mesenchymal stem cells viability in stressful microenvironments. Cell Stress Chaperone. 2015;20(2):237–51.

    Article  CAS  Google Scholar 

  3. Rahmatizadeh F, Aziz SG-G, Khodadadi K, Ataei ML, Ebrahimie E, Rad JS, et al. Bidirectional and opposite effects of naïve mesenchymal stem cells on tumor growth and progression. Adv Pharm Bull. 2019;9(4):539.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Terraza-Aguirre C, Campos-Mora M, Elizondo-Vega R, Contreras-López RA, Luz-Crawford P, Jorgensen C, et al. Mechanisms behind the immunoregulatory dialogue between mesenchymal stem cells and Th17 cells. Cells. 2020;9(7):1660.

    Article  CAS  PubMed Central  Google Scholar 

  5. Ahmad T, Mukherjee S, Pattnaik B, Kumar M, Singh S, Kumar M, et al. Miro1 regulates intercellular mitochondrial transport & enhances mesenchymal stem cell rescue efficacy. EMBO J. 2014;33(9):994–1010.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Figeac F, Lesault PF, Le Coz O, Damy T, Souktani R, Trébeau C, et al. Nanotubular crosstalk with distressed cardiomyocytes stimulates the paracrine repair function of mesenchymal stem cells. Stem Cells. 2014;32(1):216–30.

    Article  CAS  PubMed  Google Scholar 

  7. Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, et al. Mitochondrial transfer from bone-marrow–derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sinclair KA, Yerkovich ST, Hopkins PM-A, Chambers DC. Characterization of intercellular communication and mitochondrial donation by mesenchymal stromal cells derived from the human lung. Stem Cell Res Ther. 2016;7(1):91.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Spees JL, Lee RH, Gregory CA. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res Ther. 2016;7(1):125.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Asgarpour K, Shojaei Z, Amiri F, Ai J, Mahjoubin-Tehran M, Ghasemi F, et al. Exosomal microRNAs derived from mesenchymal stem cells: cell-to-cell messages. Cell Commun Signal. 2020;18(1):1–16.

    Article  Google Scholar 

  11. Kumar P, Kandoi S, Misra R, Vijayalakshmi S, Rajagopal K, Verma RS. The mesenchymal stem cell secretome: a new paradigm towards cell-free therapeutic mode in regenerative medicine. Cytokine Growth Factor Rev. 2019;46:1–9.

    Article  Google Scholar 

  12. Abbasi-Malati Z, Roushandeh AM, Kuwahara Y, Roudkenar MH. Mesenchymal stem cells on horizon: a new arsenal of therapeutic agents. Stem Cell Rev Rep. 2018;14(4):484–99.

    Article  CAS  PubMed  Google Scholar 

  13. Amiri F, Molaei S, Bahadori M, Nasiri F, Deyhim MR, Jalili MA, et al. Autophagy-modulated human bone marrow-derived mesenchymal stem cells accelerate liver restoration in mouse models of acute liver failure. Iran Biomed J. 2016;20(3):135.

    PubMed  PubMed Central  Google Scholar 

  14. Amiri F, Halabian R, Salimian M, Shokrgozar MA, Soleimani M, Jahanian-Najafabadi A, et al. Induction of multipotency in umbilical cord-derived mesenchymal stem cells cultivated under suspension conditions. Cell Stress Chaperones. 2014;19(5):657–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zhaleh F, Amiri F, Mohammadzadeh-Vardin M, Bahadori M, Harati MD, Roudkenar MH, et al. Nuclear factor erythroid-2 related factor 2 overexpressed mesenchymal stem cells transplantation, improves renal function, decreases injuries markers and increases repair markers in glycerol-induced Acute kidney injury rats. Iran J Basic Med Sci. 2016;19(3):323.

    PubMed  PubMed Central  Google Scholar 

  16. Ridge SM, Sullivan FJ, Glynn SA. Mesenchymal stem cells: key players in cancer progression. Mol Cancer. 2017;16(1):1–10.

    Article  Google Scholar 

  17. Han J, Kim B, Shin J-Y, Ryu S, Noh M, Woo J, et al. Iron oxide nanoparticle-mediated development of cellular gap junction crosstalk to improve mesenchymal stem cells’ therapeutic efficacy for myocardial infarction. ACS Nano. 2015;9(3):2805–19.

    Article  CAS  PubMed  Google Scholar 

  18. Wang M, Zhang G, Wang Y, Liu T, Zhang Y, An Y, et al. Crosstalk of mesenchymal stem cells and macrophages promotes cardiac muscle repair. Int J Biochem Cell Biol. 2015;58:53–61.

    Article  CAS  PubMed  Google Scholar 

  19. Bergfeld SA, DeClerck YA. Bone marrow-derived mesenchymal stem cells and the tumor microenvironment. Cancer Metastasis Rev. 2010;29(2):249–61.

    Article  PubMed  Google Scholar 

  20. Ahmed ES, Ahmed NH, Medhat AM, Said UZ, Rashed LA, Abdel Ghaffar ARB. Mesenchymal stem cells targeting PI3K/AKT pathway in leukemic model. Tumour Biol. 2019;41(4):1010428319846803.

    Article  CAS  PubMed  Google Scholar 

  21. Harati MD, Amiri F, Jaleh F, Mehdipour A, Harati MD, Molaee S, et al. Targeting delivery of lipocalin 2-engineered mesenchymal stem cells to colon cancer in order to inhibit liver metastasis in nude mice. Tumour Biol. 2015;36(8):6011–8.

    Article  CAS  PubMed  Google Scholar 

  22. Behrmann L, Wellbrock J, Fiedler W. The bone marrow stromal niche: a therapeutic target of hematological myeloid malignancies. Expert Opin Ther Targets. 2020;24(5):451–62.

    Article  CAS  PubMed  Google Scholar 

  23. Ciciarello M, Corradi G, Loscocco F, Visani G, Monaco F, Curti A, et al. The Yin and Yang of the bone marrow microenvironment: pros and cons of mesenchymal stromal cells in acute myeloid leukemia. Front Oncol. 2019;9:1135.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Garcia-Gomez A, De Las RJ, Ocio EM, Díaz-Rodríguez E, Montero JC, Martín M, et al. Transcriptomic profile induced in bone marrow mesenchymal stromal cells after interaction with multiple myeloma cells: implications in myeloma progression and myeloma bone disease. Oncotarget. 2014;5(18):8284.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Low JH, Ramdas P, Radhakrishnan AK. Modulatory effects of mesenchymal stem cells on leucocytes and leukemic cells: a double-edged sword? Blood Cells Mol Dis. 2015;55(4):351–7.

    Article  CAS  PubMed  Google Scholar 

  26. Ten Hacken E, Burger JA. Microenvironment interactions and B-cell receptor signaling in chronic lymphocytic leukemia: implications for disease pathogenesis and treatment. Biochim Biophys Acta. 2016;1863(3):401–13.

    Article  PubMed  Google Scholar 

  27. Zambetti NA, Ping Z, Chen S, Kenswil KJ, Mylona MA, Sanders MA, et al. Mesenchymal inflammation drives genotoxic stress in hematopoietic stem cells and predicts disease evolution in human pre-leukemia. Cell Stem Cell. 2016;19(5):613–27.

    Article  CAS  PubMed  Google Scholar 

  28. Gerdes H-H, Rustom A, Wang X. Tunneling nanotubes, an emerging intercellular communication route in development. Mech Dev. 2013;130(6–8):381–7.

    Article  CAS  PubMed  Google Scholar 

  29. Pasquier J, Guerrouahen BS, Al Thawadi H, Ghiabi P, Maleki M, Abu-Kaoud N, et al. Preferential transfer of mitochondria from endothelial to cancer cells through tunneling nanotubes modulates chemoresistance. J Transl Med. 2013;11(1):1–14.

    Article  Google Scholar 

  30. Pinto G, Brou C, Zurzolo CJ. Tunneling nanotubes: the fuel of tumor progression? Trends Cancer. 2020;6:874–88.

    Article  CAS  PubMed  Google Scholar 

  31. Kolba MD, Dudka W, Zaręba-Kozioł M, Kominek A, Ronchi P, Turos L, et al. Tunneling nanotube-mediated intercellular vesicle and protein transfer in the stroma-provided imatinib resistance in chronic myeloid leukemia cells. Cell Death Dis. 2019;10(11):1–16.

    Article  CAS  Google Scholar 

  32. Griessinger E, Moschoi R, Biondani G, Peyron J-F. Mitochondrial transfer in the leukemia microenvironment. Trends Cancer. 2017;3(12):828–39.

    Article  CAS  PubMed  Google Scholar 

  33. Matula Z, Németh A, Lőrincz P, Szepesi A, Brózik A, Buzás EI, et al. The role of extracellular vesicle and tunneling nanotube-mediated intercellular cross-talk between mesenchymal stem cells and human peripheral T cells. Stem Cell Dev. 2016;25(23):1818–32.

    Article  CAS  Google Scholar 

  34. de Rooij B, Polak R, Stalpers F, Pieters R, Den Boer M. Tunneling nanotubes facilitate autophagosome transfer in the leukemic niche. Leukemia. 2017;31(7):1651–4.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Polak R, de Rooij B, Pieters R, den Boer ML. B-cell precursor acute lymphoblastic leukemia cells use tunneling nanotubes to orchestrate their microenvironment. Blood. 2015;126(21):2404–14.

    Article  CAS  PubMed  Google Scholar 

  36. Wang J, Liu X, Qiu Y, Shi Y, Cai J, Wang B, et al. Cell adhesion-mediated mitochondria transfer contributes to mesenchymal stem cell-induced chemoresistance on T cell acute lymphoblastic leukemia cells. J Hematol Oncol. 2018;11(1):11.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Burt R, Dey A, Aref S, Aguiar M, Akarca A, Bailey K, et al. Activated stromal cells transfer mitochondria to rescue acute lymphoblastic leukemia cells from oxidative stress. Blood. 2019;134(17):1415–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kumar R, Godavarthy PS, Krause DS. The bone marrow microenvironment in health and disease at a glance. J Cell Sci. 2018;131(4):jcs201707.

    Article  PubMed  Google Scholar 

  39. Forte D, Krause DS, Andreeff M, Bonnet D, Méndez-Ferrer S. Updates on the hematologic tumor microenvironment and its therapeutic targeting. Haematologica. 2019;104(10):1928–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Moschoi R, Imbert V, Nebout M, Chiche J, Mary D, Prebet T, et al. Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid leukemic cells during chemotherapy. Blood. 2016;128(2):253–64.

    Article  CAS  PubMed  Google Scholar 

  41. Marlein C. Intercellular mitochondrial transfer in the bone marrow microenvironment of acute myeloid leukaemia and multiple myeloma. Norwich: University of East Anglia; 2018.

    Google Scholar 

  42. Marlein CR, Zaitseva L, Piddock RE, Robinson SD, Edwards DR, Shafat MS, et al. NADPH oxidase-2 derived superoxide drives mitochondrial transfer from bone marrow stromal cells to leukemic blasts. Blood. 2017;130(14):1649–60.

    Article  CAS  PubMed  Google Scholar 

  43. Omsland M, Andresen V, Gullaksen SE, Ayuda-Durán P, Popa M, Hovland R, et al. Tyrosine kinase inhibitors and interferon-α increase tunneling nanotube (TNT) formation and cell adhesion in chronic myeloid leukemia (CML) cell lines. FASEB J. 2020;34:3773–91.

    Article  CAS  PubMed  Google Scholar 

  44. Kolba MD, Dudka W, Zaręba-Kozioł M, Kominek A, Ronchi P, Turos L, et al. Tunneling nanotubes contribute to the stroma-mediated imatinib resistance of leukemic cells. bioRxiv. 2018;10.

  45. Lou E, Sperduto W, Subramanian S. Exosomes and Tunneling Nanotube Conduits: Synergistic Interaction That Facilitates Intercellular Communication Between Malignant and Stromal Cells in the Tumor Microenvironment. Diagnostic and Therapeutic Applications of Exosomes in Cancer. Amsterdam: Elsevier; 2018. p. 219–34.

    Google Scholar 

  46. Mangolini M, Ringshausen IJ. Bone marrow stromal cells drive key hallmarks of B cell malignancies. Int J Mol Sci. 2020;21(4):1466.

    Article  CAS  PubMed Central  Google Scholar 

  47. de Rooij B, Polak R, van den Berk LC, Stalpers F, Pieters R, den Boer ML. Acute lymphoblastic leukemia cells create a leukemic niche without affecting the CXCR4/CXCL12 axis. Haemetologica. 2017;102(10):e389.

    Article  Google Scholar 

  48. Beneforti L, Dander E, Bresolin S, Bueno C, Acunzo D, Bertagna M, et al. Pro-inflammatory cytokines favor the emergence of ETV6-RUNX1-positive pre-leukemic cells in a model of mesenchymal niche. Br J Haematol. 2020;190(2):262–73.

    Article  CAS  PubMed  Google Scholar 

  49. Medyouf H, Mossner M, Jann J-C, Nolte F, Raffel S, Herrmann C, et al. Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit. Cell Stem Cell. 2014;14(6):824–37.

    Article  CAS  PubMed  Google Scholar 

  50. Singh AK, Cancelas JA. Gap junctions in the bone marrow lympho-hematopoietic stem cell niche, leukemia progression, and chemoresistance. Int J Mol Sci. 2020;21(3):796.

    Article  CAS  PubMed Central  Google Scholar 

  51. Zhang X, Sun Y, Wang Z, Huang Z, Li B, Fu J. Up-regulation of connexin-43 expression in bone marrow mesenchymal stem cells plays a crucial role in adhesion and migration of multiple myeloma cells. Leuk Lymphoma. 2015;56(1):211–8.

    Article  CAS  PubMed  Google Scholar 

  52. Weber MC, Tykocinski ML. Bone marrow stromal cell blockade of human leukemic cell differentiation. Blood. 1994;83(8):2221–9.

    Article  CAS  PubMed  Google Scholar 

  53. Kouzi F, Zibara K, Bourgeais J, Picou F, Gallay N, Brossaud J, et al. Correction: Disruption of gap junctions attenuates acute myeloid leukemia chemoresistance induced by bone marrow mesenchymal stromal cells. Oncogene. 2020;39(10):2227.

    Article  CAS  PubMed  Google Scholar 

  54. Valdebenito S, Lou E, Baldoni J, Okafo G, Eugenin E. The novel roles of connexin channels and tunneling nanotubes in cancer pathogenesis. Int J Mol Sci. 2018;19(5):1270.

    Article  PubMed Central  Google Scholar 

  55. Bonacquisti EE, Nguyen J. Connexin 43 (Cx43) in cancer: implications for therapeutic approaches via gap junctions. Cancer Lett. 2019;442:439–44.

    Article  CAS  PubMed  Google Scholar 

  56. Foss B, Tronstad KJ, Bruserud Ø. Connexin-based signaling in acute myelogenous leukemia (AML). Biochim Biophys Acta. 2010;1798(1):1–8.

    Article  CAS  PubMed  Google Scholar 

  57. Fu J. Cx43 expressed on bone marrow stromal cells plays an essential role in multiple myeloma cell survival and drug resistance. Arch Med Sci. 2017;13(1):236.

    Article  CAS  PubMed  Google Scholar 

  58. Aqmasheh S. Effects of mesenchymal stem cell derivatives on hematopoiesis and hematopoietic stem cells. Adv Pharma Bull. 2017;7(2):165.

    Article  CAS  Google Scholar 

  59. Khodadi E, Shahrabi S, Shahjahani M, Azandeh S, Saki N. Role of stem cell factor in the placental niche. Cell Tissue Res. 2016;366(3):523–31.

    Article  CAS  PubMed  Google Scholar 

  60. Farahbakhshian E, Verstegen MM, Visser TP, Kheradmandkia S, Geerts D, Arshad S, et al. Angiopoietin-like protein 3 promotes preservation of stemness during ex vivo expansion of murine hematopoietic stem cells. PLoS ONE. 2014;9(8):e105642.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Oubari F, Amirizade N, Mohammadpour H, Nakhlestani M, Zarif MN. The important role of FLT3-L in ex vivo expansion of hematopoietic stem cells following co-culture with mesenchymal stem cells. Cell J (Yakhteh). 2015;17(2):201.

    Google Scholar 

  62. Richter J, Traver D, Willert K. The role of Wnt signaling in hematopoietic stem cell development. Crit Rev Biochem Mol Biol. 2017;52(4):414–24.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Huang J, Nguyen-McCarty M, Hexner EO, Danet-Desnoyers G, Klein PS. Maintenance of hematopoietic stem cells through regulation of Wnt and mTOR pathways. Nat Med. 2012;18(12):1778–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Duchartre Y, Kim Y-M, Kahn M. The Wnt signaling pathway in cancer. Crit Rev Oncol Hematol. 2016;99:141–9.

    Article  PubMed  Google Scholar 

  65. Rees WD, Sly LM, Steiner TS. How do immune and mesenchymal cells influence the intestinal epithelial cell compartment in inflammatory bowel disease? Let’s crosstalk about it! J Leukoc Biol. 2020;108:309–21.

    Article  CAS  PubMed  Google Scholar 

  66. Saki N, Abroun S, Hagh MF, Asgharei F. Neoplastic bone marrow niche: hematopoietic and mesenchymal stem cells. Cell J (Yakhteh). 2011;13(3):131.

    CAS  Google Scholar 

  67. Melzer C, von der Ohe J, Hass R. Concise review: crosstalk of mesenchymal stroma/stem-like cells with cancer cells provides therapeutic potential. Stem Cells. 2018;36(7):951–68.

    Article  CAS  PubMed  Google Scholar 

  68. Shojaei S, Hashemi SM, Ghanbarian H, Salehi M, Mohammadi-Yeganeh S. Effect of mesenchymal stem cells-derived exosomes on tumor microenvironment: tumor progression versus tumor suppression. J Cell Physiol. 2019;234(4):3394–409.

    Article  CAS  PubMed  Google Scholar 

  69. Spath C, Schlegel F, Leontyev S, Mohr FW, Dhein S. Inverse relationship between tumor proliferation markers and connexin expression in a malignant cardiac tumor originating from mesenchymal stem cell engineered tissue in a rat in vivo model. Front Pharmacol. 2013;4:42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Giallongo C, Tibullo D, Parrinello NL, La Cava P, Di Rosa M, Bramanti V, et al. Granulocyte-like myeloid derived suppressor cells (G-MDSC) are increased in multiple myeloma and are driven by dysfunctional mesenchymal stem cells (MSC). Oncotarget. 2016;7(52):85764.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Cominal JG, da Costa CM, Pinto-Simões B, Kolb H-J, Malmegrim KCR, de Castro FA. Emerging role of mesenchymal stromal cell-derived extracellular vesicles in pathogenesis of haematological malignancies. Stem Cells Int. 2019;2019:1–12.

    Article  Google Scholar 

  72. Xu S, De Veirman K, De Becker A, Vanderkerken K, Van Riet I. Mesenchymal stem cells in multiple myeloma: a therapeutical tool or target? Leukemia. 2018;32(7):1500–14.

    Article  PubMed  PubMed Central  Google Scholar 

  73. de Alvarenga EC, Silva WN, Vasconcellos R, Paredes-Gamero EJ, Mintz A, Birbrair A. Promyelocytic leukemia protein in mesenchymal stem cells is essential for leukemia progression. Ann Hematol. 2018;97(10):1749–55.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Kumar A, Anand T, Bhattacharyya J, Sharma A, Jaganathan BG. K562 chronic myeloid leukemia cells modify osteogenic differentiation and gene expression of bone marrow stromal cells. J Cell Commun Signal. 2018;12(2):441–50.

    Article  PubMed  Google Scholar 

  75. Sircar A, Chowdhury SM, Hart A, Bell WC, Singh S, Sehgal L, et al. Impact and Intricacies of bone marrow microenvironment in B-cell lymphomas: from biology to therapy. Int J Mol Sci. 2020;21(3):904.

    Article  CAS  PubMed Central  Google Scholar 

  76. Timaner M, Tsai KK, Shaked Y. The multifaceted role of mesenchymal stem cells in cancer. Semin Cancer Biol. 2020;60:225–37.

    Article  CAS  PubMed  Google Scholar 

  77. Audrito V, Vaisitti T, Serra S, Bologna C, Brusa D, Malavasi F, et al. Targeting the microenvironment in chronic lymphocytic leukemia offers novel therapeutic options. Cancer Lett. 2013;328(1):27–35.

    Article  CAS  PubMed  Google Scholar 

  78. Barcellos-de-Souza P, Gori V, Bambi F, Chiarugi P. Tumor microenvironment: bone marrow-mesenchymal stem cells as key players. Biochim Biophys Acta. 2013;1836(2):321–35.

    CAS  PubMed  Google Scholar 

  79. Ding W, Knox TR, Tschumper RC, Wu W, Schwager SM, Boysen JC, et al. Platelet-derived growth factor (PDGF)–PDGF receptor interaction activates bone marrow–derived mesenchymal stromal cells derived from chronic lymphocytic leukemia: implications for an angiogenic switch. Blood. 2010;116(16):2984–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Giannoni P, Scaglione S, Quarto R, Narcisi R, Parodi M, Balleari E, et al. An interaction between hepatocyte growth factor and its receptor (c-MET) prolongs the survival of chronic lymphocytic leukemic cells through STAT3 phosphorylation: a potential role of mesenchymal cells in the disease. Haematologica. 2011;96(7):1015–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Sharma M, Ross C, Srivastava S. Ally to adversary: mesenchymal stem cells and their transformation in leukaemia. Cancer Cell Int. 2019;19(1):1–9.

    Article  Google Scholar 

  82. Lee G-Y, Jeong S-Y, Lee H-R, Oh I-H. Age-related differences in the bone marrow stem cell niche generate specialized microenvironments for the distinct regulation of normal hematopoietic and leukemia stem cells. Sci Rep. 2019;9(1):1–12.

    Google Scholar 

  83. Agarwal P, Isringhausen S, Li H, Paterson AJ, He J, Gomariz Á, et al. Mesenchymal niche-specific expression of Cxcl12 controls quiescence of treatment-resistant leukemia stem cells. Cell Stem Cell. 2019;24(5):769-784.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Cheng J, Li Y, Liu S, Jiang Y, Ma J, Wan L, et al. CXCL8 derived from mesenchymal stromal cells supports survival and proliferation of acute myeloid leukemia cells through the PI3K/AKT pathway. FASEB J. 2019;33(4):4755–64.

    Article  CAS  PubMed  Google Scholar 

  85. Sharma M, Ross C, Srivastava S. Ally to adversary: mesenchymal stem cells and their transformation in leukaemia. Cancer Cell Int. 2019;19(1):139.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Zhou H-S, Carter BZ, Andreeff M. Bone marrow niche-mediated survival of leukemia stem cells in acute myeloid leukemia: Yin and Yang. Cancer Biol Med. 2016;13(2):248.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Ben-Batalla I, Schultze A, Wroblewski M, Erdmann R, Heuser M, Waizenegger JS, et al. Axl, a prognostic and therapeutic target in acute myeloid leukemia mediates paracrine crosstalk of leukemia cells with bone marrow stroma. Blood. 2013;122(14):2443–52.

    Article  CAS  PubMed  Google Scholar 

  88. Ma Z, Zhao X, Deng M, Huang Z, Wang J, Wu Y, et al. Bone marrow mesenchymal stromal cell-derived periostin promotes B-ALL progression by modulating CCL2 in leukemia cells. Cell Rep. 2019;26(6):1533–43.

    Article  CAS  PubMed  Google Scholar 

  89. Yu Z, Liu L, Shu Q, Li D, Wang R. Leukemia stem cells promote chemoresistance by inducing downregulation of lumican in mesenchymal stem cells. Oncol Lett. 2019;18(4):4317–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Battula VL, Le PM, Sun JC, Nguyen K, Yuan B, Zhou X, et al. AML-induced osteogenic differentiation in mesenchymal stromal cells supports leukemia growth. JCI Insight. 2017;2(13): e90036.

  91. Lee MW, Ryu S, Kim DS, Lee JW, Sung KW, Koo HH, et al. Mesenchymal stem cells in suppression or progression of hematologic malignancy: current status and challenges. Leukemia. 2019;33(3):597–611.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Liang W, Chen X, Zhang S, Fang J, Chen M, Xu Y, et al. Mesenchymal stem cells as a double-edged sword in tumor growth: focusing on MSC-derived cytokines. Cell Mol Biol Lett. 2021;26(1):1–25.

    Article  Google Scholar 

  93. Vianello F, Villanova F, Tisato V, Lymperi S, Ho K-K, Gomes AR, et al. Bone marrow mesenchymal stromal cells non-selectively protect chronic myeloid leukemia cells from imatinib-induced apoptosis via the CXCR4/CXCL12 axis. Haematologica. 2010;95(7):1081–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Stamatopoulos B, Meuleman N, De Bruyn C, Pieters K, Mineur P, Le Roy C, et al. AMD3100 disrupts the cross-talk between chronic lymphocytic leukemia cells and a mesenchymal stromal or nurse-like cell-based microenvironment: pre-clinical evidence for its association with chronic lymphocytic leukemia treatments. Haematologica. 2012;97(4):608–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Kojima K, McQueen T, Chen Y, Jacamo R, Konopleva M, Shinojima N, et al. p53 activation of mesenchymal stromal cells partially abrogates microenvironment-mediated resistance to FLT3 inhibition in AML through HIF-1α–mediated down-regulation of CXCL12. Blood. 2011;118(16):4431–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications. Mol Ther. 2015;23(5):812–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Nawaz M, Fatima F, Vallabhaneni KC, Penfornis P, Valadi H, Ekström K, et al. Extracellular vesicles: evolving factors in stem cell biology. Stem Cells Int. 2016;2016:1–17.

    Article  Google Scholar 

  98. Laurenzana I, Lamorte D, Trino S, De Luca L, Ambrosino C, Zoppoli P, et al. Extracellular vesicles: a new prospective in crosstalk between microenvironment and stem cells in hematological malignancies. Stem Cells Int. 2018;2018:1–11.

    Article  Google Scholar 

  99. Muntión S, Ramos TL, Diez-Campelo M, Rosón B, Sánchez-Abarca LI, Misiewicz-Krzeminska I, et al. Microvesicles from mesenchymal stromal cells are involved in HPC-microenvironment crosstalk in myelodysplastic patients. PLoS ONE. 2016;11(2):e0146722.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Kordelas L, Rebmann V, Ludwig A, Radtke S, Ruesing J, Doeppner T, et al. MSC-derived exosomes: a novel tool to treat therapy-refractory graft-versus-host disease. Leukemia. 2014;28(4):970–3.

    Article  CAS  PubMed  Google Scholar 

  101. Del Fattore A, Luciano R, Pascucci L, Goffredo BM, Giorda E, Scapaticci M, et al. Immunoregulatory effects of mesenchymal stem cell-derived extracellular vesicles on T lymphocytes. Cell Transplant. 2015;24(12):2615–27.

    Article  PubMed  Google Scholar 

  102. Favaro E, Carpanetto A, Lamorte S, Fusco A, Caorsi C, Deregibus MC, et al. Human mesenchymal stem cell-derived microvesicles modulate T cell response to islet antigen glutamic acid decarboxylase in patients with type 1 diabetes. Diabetologia. 2014;57(8):1664–73.

    Article  CAS  PubMed  Google Scholar 

  103. Mineo M, Garfield SH, Taverna S, Flugy A, De Leo G, Alessandro R, et al. Exosomes released by K562 chronic myeloid leukemia cells promote angiogenesis in a Src-dependent fashion. Angiogenesis. 2012;15(1):33–45.

    Article  CAS  PubMed  Google Scholar 

  104. Taverna S, Flugy A, Saieva L, Kohn EC, Santoro A, Meraviglia S, et al. Role of exosomes released by chronic myelogenous leukemia cells in angiogenesis. Int J Cancer. 2012;130(9):2033–43.

    Article  CAS  PubMed  Google Scholar 

  105. Wang J, Faict S, Maes K, De Bruyne E, Van Valckenborgh E, Schots R, et al. Extracellular vesicle cross-talk in the bone marrow microenvironment: implications in multiple myeloma. Oncotarget. 2016;7(25):38927.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Crompot E, Van Damme M, Pieters K, Vermeersch M, Perez-Morga D, Mineur P, et al. Extracellular vesicles of bone marrow stromal cells rescue chronic lymphocytic leukemia B cells from apoptosis, enhance their migration and induce gene expression modifications. Haematologica. 2017;102(9):1594–604.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Jiang Y-H, Liu J, Lin J, Li S-Q, Xu Y-M, Min Q-H, et al. K562 cell-derived exosomes suppress the adhesive function of bone marrow mesenchymal stem cells via delivery of miR-711. Biochem Biophys Res Commun. 2020;521(3):584–9.

    Article  CAS  PubMed  Google Scholar 

  108. Zhang F, Lu Y, Wang M, Zhu J, Li J, Zhang P, et al. Exosomes derived from human bone marrow mesenchymal stem cells transfer miR-222-3p to suppress acute myeloid leukemia cell proliferation by targeting IRF2/INPP4B. Mol Cell Probes. 2020;51:101513.

    Article  CAS  PubMed  Google Scholar 

  109. Zhang F, Li J, Zhu J, Liu L, Zhu K, Cheng S, et al. IRF2–INPP4B-mediated autophagy suppresses apoptosis in acute myeloid leukemia cells. Biol Res. 2019;52(1):11.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Gruszka AM, Valli D, Restelli C, Alcalay M. Adhesion deregulation in acute myeloid leukaemia. Cells. 2019;8(1):66.

    Article  CAS  PubMed Central  Google Scholar 

  111. Whiteside TL. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin Immunol. 2018;35:69–79.

    Article  CAS  PubMed  Google Scholar 

  112. Dostert G, Mesure B, Menu P, Velot É. How do mesenchymal stem cells influence or are influenced by microenvironment through extracellular vesicles communication? Front Cell Dev Biol. 2017;5:6.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Lopatina T, Gai C, Deregibus MC, Kholia S, Camussi G. Cross talk between cancer and mesenchymal stem cells through extracellular vesicles carrying nucleic acids. Front Oncol. 2016;6:125.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Roccaro AM, Sacco A, Maiso P, Azab AK, Tai Y-T, Reagan M, et al. BM mesenchymal stromal cell–derived exosomes facilitate multiple myeloma progression. J Clin Investig. 2013;123(4):1542–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Cheng Q, Li X, Liu J, Ye Q, Chen Y, Tan S, et al. Multiple myeloma-derived exosomes regulate the functions of mesenchymal stem cells partially via modulating miR-21 and miR-146a. Stem Cells Int. 2017;2017:1–9.

    Article  Google Scholar 

  116. De Luca L, Laurenzana I, Trino S, Lamorte D, Caivano A, Musto P. An update on extracellular vesicles in multiple myeloma: a focus on their role in cell-to-cell cross-talk and as potential liquid biopsy biomarkers. Expert Rev Mol Diagn. 2019;19(3):249–58.

    Article  PubMed  Google Scholar 

  117. Trimarco V, Ave E, Facco M, Chiodin G, Frezzato F, Martini V, et al. Cross-talk between chronic lymphocytic leukemia (CLL) tumor B cells and mesenchymal stromal cells (MSCs): implications for neoplastic cell survival. Oncotarget. 2015;6(39):42130.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Yang H, Tabe Y, Saito K, Yamatani K, Jacamo R, Ma H, et al. Oxphos inhibition induces formation of tunneling nanotubes in AML cells and facilitates mitochondrial transfer from BM stroma to AML that contributes to microenvironment-mediated drug-resistance of AML. Washington, DC: American Society of Hematology; 2019.

    Book  Google Scholar 

  119. Omsland M, Bruserud Ø, Gjertsen BT, Andresen V. Tunneling nanotube (TNT) formation is downregulated by cytarabine and NF-κB inhibition in acute myeloid leukemia (AML). Oncotarget. 2017;8(5):7946.

    Article  PubMed  Google Scholar 

  120. Zhang Y, Hu K, Hu Y, Liu L, Wang B, Huang H. Bone marrow mesenchymal stromal cells affect the cell cycle arrest effect of genotoxic agents on acute lymphocytic leukemia cells via p21 down-regulation. Ann Hematol. 2014;93(9):1499–508.

    Article  CAS  PubMed  Google Scholar 

  121. Xia C, Wang T, Cheng H, Dong Y, Weng Q, Sun G, et al. Mesenchymal stem cells suppress leukemia via macrophage-mediated functional restoration of bone marrow microenvironment. Leukemia. 2020;34:1–9.

    Article  Google Scholar 

  122. Jackson MV, Krasnodembskaya AD. Analysis of mitochondrial transfer in direct co-cultures of human monocyte-derived macrophages (MDM) and mesenchymal stem cells (MSC). Bio Protoc. 2017;7(9): e2255.

  123. Wu Y, Campos L, Daguenet E, He Z, Picot T, Tavernier-Tardy E, et al. FAK deficiency in bone marrow stromal cells alters their homeostasis and drives abnormal proliferation and differentiation of haematopoietic stem cells. Cells. 2020;9(3):646.

    Article  CAS  PubMed Central  Google Scholar 

  124. Ito S, Barrett AJ, Dutra A, Pak E, Miner S, Keyvanfar K, et al. Long term maintenance of myeloid leukemic stem cells cultured with unrelated human mesenchymal stromal cells. Stem Cell Res. 2015;14(1):95–104.

    Article  CAS  PubMed  Google Scholar 

  125. Brenner AK, Nepstad I, Bruserud Ø. Mesenchymal stem cells support survival and proliferation of primary human acute myeloid leukemia cells through heterogeneous molecular mechanisms. Front Immunol. 2017;8:106.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Xu S, Menu E, Becker AD, Van Camp B, Vanderkerken K, Van Riet I. Bone marrow-derived mesenchymal stromal cells are attracted by multiple myeloma cell-produced chemokine CCL25 and favor myeloma cell growth in vitro and in vivo. Stem Cells. 2012;30(2):266–79.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was confirmed and founded by Vice-chancellor for Research and Technology, Hamadan University of Medical Sciences, Ethic Code: IR.UMSHA.REC.1400.184.

Funding

This work was founded by the Hamadan University of Medical Sciences (Grant No. 140004012855).

Author information

Authors and Affiliations

Authors

Contributions

MV and AS have participated to write the first manuscript. FA and AG have revised and completed some shortcomings to improve it. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Fatemeh Amiri.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors contributed in this review clearly state that there is no conflict of interest either in authorship or financial benefits.

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 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Goodarzi, A., Valikhani, M., Amiri, F. et al. The mechanisms of mutual relationship between malignant hematologic cells and mesenchymal stem cells: Does it contradict the nursing role of mesenchymal stem cells?. Cell Commun Signal 20, 21 (2022). https://doi.org/10.1186/s12964-022-00822-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-022-00822-6

Keywords