Background: T-cell acute lymphoblastic leukemia (T-ALL) is a malignant hematologic disease caused by the transformation and uncontrolled proliferation of T-cell precursors. T-ALL is generally thought to originate in the thymus since lymphoblasts express phenotypic markers comparable to those described in thymocytes in distinct stages of development. Although around 50% of T-ALL patients present a thymic mass, T-ALL is characterized by peripheral blood and bone marrow involvement, and central nervous system (CNS) infiltration is one of the most severe complications of the disease. Summary: The CNS invasion is related to the expression of specific adhesion molecules and receptors commonly expressed in developing T cells, such as L-selectin, CD44, integrins, and chemokine receptors. Furthermore, T-ALL blasts also express neurotransmitters, neuropeptides, and cognate receptors that are usually present in the CNS and can affect both the brain and thymus, participating in the crosstalk between the organs. Key Messages: This review discusses how the thymus-brain connections, mediated by innervation and common molecules and receptors, can impact the development and migration of T-ALL blasts, including CNS infiltration.

T-cell acute lymphoblastic leukemia (T-ALL) is a neoplasm of lymphoblasts committed to the T-cell lineage. It is currently classified as a form of T-cell lymphoblastic leukemia/lymphoma (T-ALL/LBL). T-ALL involves peripheral blood and bone marrow infiltration. In contrast, T-cell lymphoblastic lymphoma (T-LBL) presents with primary involvement of the thymus, nodal, or extranodal sites, with no or minimal evidence of peripheral blood and bone marrow involvement [1]. Thus, T-LBL and T-ALL are distinguished based on a 20% bone marrow infiltration cutoff point. When bone marrow infiltration levels are below 20%, it characterizes T-LBL, whereas levels above this threshold are classified as T-ALL [2].

T-ALL accounts for about 15% of childhood and 25% of adult ALL cases. It is still considered a high-risk disease associated with a higher risk of induction failure, early relapse, and central nervous system (CNS) infiltration and relapse compared to the most common B-cell acute lymphoblastic leukemia. Among T-ALL subtypes, the early T-cell precursor lymphoblastic leukemia (ETP-ALL) accounts for approximately 10–13% of T-ALL cases in childhood and 5–10% of adult ALL cases, presenting a unique immunophenotypic and genomic profile. This profile seems to represent a higher risk disease subtype than non-ETP-ALL in adults [1], being associated with higher initial refractoriness to treatment, higher relapse risk, and lower overall survival [3]. However, other studies have shown no difference in overall survival between ETP-ALL and T-ALL patients [4‒6].

Although the most common site of relapse of T-ALL is the bone marrow, infiltration of the CNS also occurs, which is particularly important regarding the risk of leukemic relapse and disease prognosis. Disseminated T-ALL is typically isolated to the leptomeninges and rarely invades the brain parenchyma. Among the entry routes for ALL cells in the CNS, the blood-cerebrospinal fluid barrier has been proposed to serve as a primary entry point for the blasts [7]. Indeed, CNS involvement is measured by the detection of leukemic cells in the cerebrospinal fluid (CSF) after a lumbar puncture. However, some studies have shown that B-ALL cells migrate directly from the bone marrow to the CNS, bypassing the need to enter and exit the CNS vasculature [8, 9], suggesting this possibility for T-ALL cells.

Different strategies for therapies involve the search for biomarkers and the use of drugs that target CNS invasion and survival mechanisms employed by leukemia cells in the leptomeninges [10]. Some of these drugs target adhesion molecules such as integrins (e.g., VLA-4) and chemokine receptors (e.g., CXCR4) which may be involved in the preferential location of T-ALL cells in the CSF and are also crucial for the survival, migration, and egress of T-ALL cells and normal thymocytes from the thymus [10, 11].

The thymus-brain connection can be mediated by innervation, through the release of neurotransmitters, and by hormones and other soluble molecules produced by both organs. Notably, various receptors for neurotransmitters, neuropeptides, and hormones are expressed in developing thymocytes and T-ALL blasts [12, 13]. Their activation can affect cell survival, proliferation, and migration, which are key biological responses impacting intrathymic T-cell development (for a comprehensive overview of human T-cell development, we refer to a recent review by Bosticardo and Notarangelo [14]). Conversely, lymphoblasts produce cytokines and chemokines that could have local and systemic effects on CNS activity once infiltrated, for example, activating microglia and promoting or facilitating CNS infiltration. In this review, we discuss the molecules possibly involved in the thymus-brain crosstalk in T-ALL, including those commonly expressed by the two systems, and their possible roles in blast migration and infiltration of the CNS.

Neurotransmitters, comprising amino acids, monoamines, and peptides, are constitutively produced in the thymus by both autonomic innervation and thymic cells [15, 16]. Accordingly, thymic microenvironmental cells and developing thymocytes express related receptors [12], which, upon activation, can modulate thymic physiology and thymocyte differentiation.

Thymic innervation is characterized by the presence of autonomic nervous system fibers, providing sympathetic noradrenergic fibers within the parenchyma [15, 17, 18]. The primary source of these nerves is postganglionic neurons in the superior cervical and stellate ganglia [19]. Using pseudorabies virus (PRV) to trace neural connections, a study also identified the preganglionic sympathetic neurons responsible for innervating the thymus in rats. These neurons were found in the intermediolateral cell column of the T1–T7 spinal cord segments [20]. Interestingly, this study observed considerable overlap with CNS cell groups connected to the sympathetic innervation of non-immune organs, such as the spleen and bone marrow, suggesting that all sympathetically innervated organs have some neuronal populations in common.

Nerve fibers are observed in all thymic compartments, including the capsule/subcapsular region, cortex, cortico-medullary junction, and medulla, in close association with thymocytes and thymic epithelial cells (TECs), as well as with dendritic cells, macrophages, and B cells [18]. The widespread presence of nerve fibers strongly suggests the possible link between the nervous system and all stages of T-cell development, which depends on the interaction of developing thymocytes with other cells in the thymic microenvironment. However, the direct role of thymus innervation in T-lymphocyte differentiation is still in need of further confirmation. The thymus also seems to be innervated by the parasympathetic fibers from branches of the vagus nerve [21]. However, this is still a matter of debate [16]. Nevertheless, there is an intrathymic non-neuronal cholinergic system represented by the presence of acetylcholine (ACh), which is synthesized and released by TECs and thymocytes, and the expression of the corresponding receptors by both cell compartments [22‒24].

Besides normal developing lymphocytes, T-ALL primary cells and cell lines express neurotransmitters, neuropeptides, and cognate receptors, as summarized in Table 1. This expression may influence the survival and function of T-ALL blasts, as well as the adjacent microenvironment, affecting nearby cells including CNS and other immune cells. In terms of CNS infiltration, the production of neurotransmitters by T-ALL could change normal brain function. This could potentially lead to neurological symptoms or complications in patients with T-ALL. The expression and importance of neurotransmitters and their receptors in T-ALL blasts will be discussed in more detail in the following sections.

Table 1.

Examples of molecules and receptors possibly involved in the thymus-brain connections in T-ALL

MoleculeExpression and/or functionReferences
β-ARs • Levels significantly elevated in primary T-ALL cells [25
• Inhibition of cell proliferation, induction of apoptosis, and blockage of the cell cycle, exhibiting anti-tumor effects in vitro and in vivo 
ACh and cholinergic receptors • ACh production by T-ALL cells is considerably higher when compared to B-ALL cells [26‒28
• AChRs play an important role in leukemic cell proliferation, differentiation, and apoptosis 
• mAChRs are linked to NO synthesis by nNOS and/or iNOS by lymphoblasts and endothelial cells 
Glutamate • T-ALL cell lines and CNS cells express glutamate receptor GluR3 [29‒31
• Increase of T-ALL cell lines adhesion to fibronectin 
• Increase of MMP-9 secretion by T-ALL cell lines, increasing the invasive potential of cells 
• Improves engraftment of human T-leukemia cells in vivo 
• Elevated levels of glutamate are found in the CSF of patients with ALL 
DA • Induces T-cell adhesion to fibronectin and ICAM [32‒34
• T-ALL cell lines express DA receptors 
• Genetic polymorphisms that regulate DA in primary T-ALL cells were associated with CSF biomarkers 
SEMAs and NRPs • Plasma levels of NRP1 are higher in ALL patients compared with healthy volunteers [35‒37
• T-ALL primary cells and cell lines express NRP2 
• NRP2 plays a role in T-ALL cells migration 
• NRPs are involved in neuronal development by participating in axonal guidance 
S1P/S1P1 • T-ALL cells express S1P1 and migrate toward S1P [35, 38‒42
• S1P induces chemotaxis and fugetaxis of lymphoblasts depending on the concentration 
• S1PRs are expressed in different cell types in the CNS 
• S1P facilitates glutamate secretion 
CXCL12/CXCR4 • CXCL12 is highly concentrated in the CSF. In vitro invasion of ALL cells into human CSF is partially inhibited by blocking CXCR4 [7, 8, 43‒46
• CXCR4 mediates the invasion of the meninges by T-ALL cells stimulated by CXCL12 
• CXCR4 antagonists disrupt lymphoblast adhesion to meningeal cells, reverse chemoresistance, and induce cell death 
• CXCR4 is required for T-ALL cell motility 
• Blocking CXCR4 in T-ALL cells prevents CNS infiltration in vivo 
CCL19/CCR7 • CCR7–CCL19 interactions take part in CNS infiltration in vitro and in vivo [45, 47
• Silencing CCR7 in vivo inhibits CNS infiltration 
Thymulin • Thymulin is produced in the thymus and in the brain, by astrocytes and glial cells (rodents) [48‒50
• Lower plasma levels of active thymulin have been observed in samples from patients with T-ALL 
• Thymulin reduces the proliferation of primary T-ALL and cell lines 
MoleculeExpression and/or functionReferences
β-ARs • Levels significantly elevated in primary T-ALL cells [25
• Inhibition of cell proliferation, induction of apoptosis, and blockage of the cell cycle, exhibiting anti-tumor effects in vitro and in vivo 
ACh and cholinergic receptors • ACh production by T-ALL cells is considerably higher when compared to B-ALL cells [26‒28
• AChRs play an important role in leukemic cell proliferation, differentiation, and apoptosis 
• mAChRs are linked to NO synthesis by nNOS and/or iNOS by lymphoblasts and endothelial cells 
Glutamate • T-ALL cell lines and CNS cells express glutamate receptor GluR3 [29‒31
• Increase of T-ALL cell lines adhesion to fibronectin 
• Increase of MMP-9 secretion by T-ALL cell lines, increasing the invasive potential of cells 
• Improves engraftment of human T-leukemia cells in vivo 
• Elevated levels of glutamate are found in the CSF of patients with ALL 
DA • Induces T-cell adhesion to fibronectin and ICAM [32‒34
• T-ALL cell lines express DA receptors 
• Genetic polymorphisms that regulate DA in primary T-ALL cells were associated with CSF biomarkers 
SEMAs and NRPs • Plasma levels of NRP1 are higher in ALL patients compared with healthy volunteers [35‒37
• T-ALL primary cells and cell lines express NRP2 
• NRP2 plays a role in T-ALL cells migration 
• NRPs are involved in neuronal development by participating in axonal guidance 
S1P/S1P1 • T-ALL cells express S1P1 and migrate toward S1P [35, 38‒42
• S1P induces chemotaxis and fugetaxis of lymphoblasts depending on the concentration 
• S1PRs are expressed in different cell types in the CNS 
• S1P facilitates glutamate secretion 
CXCL12/CXCR4 • CXCL12 is highly concentrated in the CSF. In vitro invasion of ALL cells into human CSF is partially inhibited by blocking CXCR4 [7, 8, 43‒46
• CXCR4 mediates the invasion of the meninges by T-ALL cells stimulated by CXCL12 
• CXCR4 antagonists disrupt lymphoblast adhesion to meningeal cells, reverse chemoresistance, and induce cell death 
• CXCR4 is required for T-ALL cell motility 
• Blocking CXCR4 in T-ALL cells prevents CNS infiltration in vivo 
CCL19/CCR7 • CCR7–CCL19 interactions take part in CNS infiltration in vitro and in vivo [45, 47
• Silencing CCR7 in vivo inhibits CNS infiltration 
Thymulin • Thymulin is produced in the thymus and in the brain, by astrocytes and glial cells (rodents) [48‒50
• Lower plasma levels of active thymulin have been observed in samples from patients with T-ALL 
• Thymulin reduces the proliferation of primary T-ALL and cell lines 

Epinephrine, Norepinephrine, and Adrenergic Receptors

Epinephrine and norepinephrine (adrenaline and noradrenaline, respectively) are hormones and neurotransmitters released from the adrenal glands, whereas norepinephrine is released from the sympathetic nerve terminals. They act through adrenergic receptors (ARs) present on many cell types, including developing T lymphocytes [51].

Different studies have found that epinephrine and norepinephrine exert stimulatory effects on cancer development and progression, and β-adrenergic receptor (β-AR) blockers have anti-tumor effects, inhibiting melanoma, rectal, breast, and gastric cancer cell proliferation [52‒55]. The expression and role of β-adrenergic receptors (β-ARs) were observed in T-ALL primary cells and cell lines such as Jurkat, CEM-C1, and CEM-C7. Xu and collaborators [25] recently showed that newly diagnosed T-ALL patients express higher levels of β1, β2, and β3-ARs than the control group. Using Carvedilol, a third-generation non-selective β-AR blocker, they observed inhibition of T-ALL viability in vitro and in vivo by the induction of apoptosis and arrest of the cell cycle. Treatment with Carvedilol reduced β1, β2, and β3-ARs levels in CEM-C1 and CEM-C7 cells and inhibited their downstream signaling pathways, reducing PI3K, p-PI3K, AKT, p-AKT, mTOR, JAK2, STAT3, and p-STAT3 levels. In vivo, Carvedilol treatment notably reduced both the size and weight of xenograft tumors in mice. Additionally, it triggered apoptosis in the tumors while reducing the presence of CEM-C1 cells in murine peripheral blood and bone marrow.

Propranolol, another non-selective β-AR blocker, induced cytotoxicity in vitro, in Jurkat and MOLT-4 cells. Also, propranolol significantly decreased vascular endothelial growth factor (VEGF) production and MMP-2 (metalloprotease-2) activity when the cell lines were activated with PMA [56, 57]. VEGF is a co-mitogen for the vascular endothelial cells, promoting vascular permeability, regeneration, and angiogenesis. The importance of angiogenic factors in solid tumors is well described, but it also seems to be correlated with the treatment, relapse, and prognosis of hematolymphoid tumors such as T-ALL [58]. It was indeed shown in B-ALL in vivo, by transplanting leukemia samples into NOD/SCID mice. CNS ALL cells were characterized by high expression of VEGF transcripts when compared with corresponding bone marrow-derived samples. Importantly, a clear reduction in meningeal infiltration was seen in animals treated with bevacizumab (which specifically binds to human but not to murine VEGF) as compared with controls. No effect of leukemia reduction was observed in the bone marrow or spleen, indicating that ALL invasion of the CNS is mediated by human ALL-derived VEGF and can be reduced by VEGF inhibition [59].

MMPs are zinc-dependent proteases involved in the degradation of extracellular matrix components, cleavage of membrane receptors such as integrins, as well as chemokine and cytokine processing, which modulate cell migration and spread [60]. Yet, whether β-AR blockers modulate T-ALL migratory patterns and invasion needs further examination. In any case, these data show the potential role of β-AR activation or inhibition in T-ALL development.

ACh and Cholinergic Receptors

As mentioned above, there is an intrathymic cholinergic system represented by ACh, which is synthesized and released by TECs and thymocytes that also express cholinergic receptors [22‒24]. Cholinergic receptors (AChRs) consist of two major subtypes: the metabotropic G-protein-coupled muscarinic receptors (mAChRs) and the ionotropic nicotinic receptors (nAChRs). Multiple nAChRs subunits have been described as being expressed by normal thymocytes and TECs (e.g., α3, α5, α7, β2, and β4 in thymocytes; and α2, α3, α5, α7, β4, and ε in TECs) [12]. The expression of mAChRs and nAChRs in thymocytes was also confirmed in binding studies using radiolabeled ligands [61].

The cholinergic machinery is also present in T-ALL blasts, and a more detailed overview of this subject has been reviewed in detail by Dobrovinskaya and colleagues [26]. ACh production by T-ALL cell lines (CEM, HSB-2, Jurkat, MOLT-3, and MOLT-4) is considerably higher when compared to B-ALL cell lines and unstimulated peripheral blood mononuclear leukocytes obtained from healthy donors [23, 27]. One of the main mechanisms responsible for appropriate cholinergic function is performed by the acetylcholinesterase (AChE) enzyme. Interestingly, a study has shown that blood and lymphocyte AChE activity was increased in the newly diagnosed T- and B-ALL samples and decreased in the remission induction group in relation to the controls, reinforcing the possible role of the cholinergic machinery in leukemia [62].

AChRs have been shown to play an important role in leukemic cell proliferation, differentiation, and apoptosis. The presence of the mRNA-encoding nAChRs α3, α5, α6, α9, and α10 subunits was reported in T-ALL cell lines as CEM and Jurkat [63, 64]. Jurkat, but not CEM, also expresses the α7 subunit [64]. Regarding mAChRs, Jurkat cells present high M3 expression, lower M4 and M5, and no M1 and M2 expression [65]. CEM cells also express functional mAChRs linked to NO synthesis by nNOS and/or iNOS [28]. Also, both mAChRs and nAChRs were described in T-ALL primary cells [66], suggesting that these cells can also respond to ACh or ACh agonists.

Since T-ALL blasts produce a large quantity of ACh, it is conceivable that it may also exert a significant paracrine effect on neighboring stromal cells in their local niche. Based on that, Dobrovinskaya and colleagues [26] suggested a hypothetical model of non-neuronal cholinergic system involvement in T leukemogenesis in the bone marrow. In this scenario, ACh produced by T-ALL blasts would induce an autocrine activation leading to proliferation and a paracrine effect on osteoblasts and endothelial cells that express nAChRs and mAChRs, playing an important role in the remodeling of microenvironmental niches. In this same line, one can argue that a similar phenomenon could occur in other niches or sites of blast invasion, such as the CNS. The concept of a molecule that influences both leukemic cells and the different niches where these cells are located may be of great interest for a better understanding of the pathogenesis of the disease. Furthermore, the role of ACh released by human T lymphocytes on endothelial cells was recently shown [67]. T-cell-derived ACh increased endothelial nitric oxide synthase activity, promoted vasorelaxation, reduced vascular endothelial activation, and promoted barrier integrity, suggesting a cholinergic immune regulation of vascular endothelial function in human inflammation. Thus, it is also conceivable that local ACh release by T-ALL cells impacts cell transmigration and access to different tissues.

Glutamate and Glutamate Receptors

Glutamate is the most abundant excitatory neurotransmitter in the CNS, involved in different aspects of the brain’s physiology and function. Glutamate acts via two classes of receptors: GluRs that are ligand-gated ion channels and called ionotropic glutamate receptors (iGluRs), and GluRs that are G-protein-coupled and called metabotropic glutamate receptors, which activate intracellular signal transduction pathways following binding of glutamate. GluRs are expressed by various immune cells, including dendritic cells, T cells, T-LBL and T-ALL blasts, and cell lines such as Jurkat and FRO [68].

Studies by Ganor et al. [29, 69] showed that Jurkat cells express high levels of iGluR3, and glutamate treatment increases the membrane expression of CD147 in these cells, which is an inducer of MMPs and could, consequently, modulate cell adhesion and migration. Employing an experimental model of leukemia engraftment by injecting Jurkat cells into chick embryos, they have shown that glutamate treatment enhanced Jurkat engraftment and pro-metastatic effect [29].

More recently, chimeric antigen receptor (CAR) T-cell therapy targeting human CD147 was used for treating T-ALL [70]. CD147-CAR T cells showed anti-tumor activity against T-ALL primary and Jurkat cells by cytotoxicity mediated by granzyme B and perforin and by releasing a high level of cytokines. CD147-CAR T cells also controlled the progression of human T-ALL in a mouse xenograft model, showing their promising potential as a target for T-ALL therapy. These data reinforce the potential role of glutamate in T-ALL since it modulates CD147 membrane expression in T-ALL cells in vivo.

Besides that, glutamate increases integrin-mediated T-cell adhesion to laminin and fibronectin and CXCR4-dependent chemotactic migration toward CXCL12 [29]. Those are also critical events for the migration and spread of T-ALL blasts. Whether glutamate increases T-ALL cell adhesion and migration and CNS infiltration still needs further investigation.

Dopamine and Dopamine Receptors

Dopamine (DA) is a catecholamine neurotransmitter extensively distributed in the CNS and involved in regulating many vital physiological processes [71, 72]. The interaction of DA with one of the five DA G-protein-coupled receptors (DR 1-5) activates a complex signaling network involved in the regulation of motor neurons, modulation of emotional states, cognitive function, and hormonal and sympathetic regulation [71, 73, 74].

DA can interact with cells from the immune system, regulating immune functions such as cell adhesion, cytokine secretion, and chemotaxis [32, 75, 76]. Mignini et al. [16] observed the presence of DA and correlated receptors in the thymus of rodents, mainly in the medulla and cortico-medullary junction. In addition, an increase in the number of thymic regulatory T cells (tTregs) was observed in conditions of prenatal catecholamine deficiency, suggesting the participation of DA and its receptors in tTreg development [77].

T-ALL cells also express DRs, and the possible role of DA in T-ALL development was suggested through in vitro experiments. Basu et al. [33] showed that Jurkat cells express DR1 and DR2, like normal T cells, but diverge in the low expression of DR3, DR4, and DR5. Specific DR1 and DR2 agonists inhibited the proliferation of normal T cells. Still, this effect was not observed in Jurkat cells due to the failure of DR1-mediated activation of cyclic AMP signaling and missense mutation of DR2 that affects the inhibition of phosphorylation of ZAP-70, an essential downstream protein transducing signal from the T-cell receptor. These results suggest the involvement of DA and alterations in DR1 and DR2 receptors in the uncontrolled proliferation of leukemic cells. In addition, polymorphisms in genes encoding enzymes involved in the production and degradation of DA and DR4 were associated with CSF biomarkers of CNS injury in ALL primary cells of children under treatment [34]. In this study, the distinction between ALL patients was not described, but 15% of patients had T-ALL.

Semaphorins and Neuropilins

Semaphorins (SEMAs) are axon-guiding glycoproteins with repulsive properties that can bind neuropilins (NRPs) and plexins, initially described in the nervous system. SEMAs are classified into eight classes: classes 1 and 2 are found in invertebrates, classes 3 to 7 comprise vertebrate SEMAs, and class V represents viral-encoded SEMAs. Vertebrate membrane-bound SEMAs (classes 4–7) bind directly to plexin subclasses A–D, whereas secreted SEMAs (class 3) bind NRPs (1 and 2). As NRPs have a short cytoplasmic tail, they bind to class A plexins as co-receptors able to activate signaling pathways. The expression of SEMAs, NRPs, and plexins is well described in the immune system. Some are found in the human thymus, expressed by TEC and thymocytes, mediating thymocyte migration and thymocyte-TEC interactions [78].

NRPs and SEMAs have been described in different pathologies, mainly neurodevelopmental disorders and cancer. The expression levels of NRPs and SEMAs are also related to the progression of solid tumors. Besides the expression of class 3 SEMAs A and F, NRP1 and NRP2, and plexin A in the human thymuses in TECs and developing thymocytes, their expression was also found in the thymus in T-ALL and T-LBL conditions. Interestingly, NRP2 and SEMA3F are highly expressed in T-LBL compared to T-ALL samples [35]. In addition, the SEMA3F protein levels in T-LBL biopsies were higher in intrathymic malignant cells when compared with the normal thymus or with T-LBL cells localized in other peripheral sites such as lymph nodes and non-lymphoid organs. Functionally, SEMA3F can inhibit T-ALL and LBL migration toward CXCL12 and sphingosine-1-phosphate (S1P), two important chemotactic factors related to blast localization and infiltration. Malignant T-cell precursors respond to SEMA3F similarly to normal thymocytes, meaning that T-ALL blasts expressing lower levels of NRP2 and SEMA3F would respond and migrate toward CXCL12 and S1P. In contrast, blasts expressing higher levels of NRP2 and SEMA3F would not respond to CXCL12 and S1P appropriately, being less able to migrate and infiltrate [35].

NRPs can also bind to different members of the VEGF family, playing an important role not only in vascular and neuronal development but also in the proliferation and migration of leukemic cells [36, 58, 79]. As discussed above, samples from patients with ALL have elevated levels of VEGF-A in the CSF compared to the serum. Importantly, elevated VEGF-A levels were observed in the CSF of patients with CNS infiltration compared with patients without CNS infiltration, suggesting the participation of VEGF in CNS metastasis [80]. Since NRPs are also receptors for VEGFs, they might participate in VEGF-mediated CNS invasion.

S1P and S1P Receptor 1

S1P is a phospholipid involved in different cell processes, including thymocyte survival, egress from the thymus, and peripheral T-lymphocyte migration and egress from peripheral lymphoid organs. Primary T-ALL and T-LBL cells and different cell lines respond to S1P through the activation of S1P1 [35, 38]. Interestingly, S1P activation can result in two different migratory responses in vitro: lower S1P concentrations induce chemotaxis, whereas higher concentrations induce the fugetaxis of cell lines such as Jurkat, CEM, MOLT-4, and HPB-ALL. The analysis of signaling pathways following low or high doses of S1P suggests that Rac1 activity is possibly involved in regulating these specific migratory responses [38]. Moreover, using a T-LBL model in zebrafish, a study has shown that the inhibition of S1P1 induces lymphoma cells to disaggregate and intravasate into the vasculature in vivo, thus implicating high S1P1 levels in the blockade of T-LBL dissemination [81].

Besides the role of S1P in T-ALL and T-LBL migration and thymocyte egress, it can also mediate neuronal signaling and function, suggesting that it could mediate the thymus-brain crosstalk. First, directly, by possibly inducing CNS infiltration of T-ALL blasts. Second, indirectly, by triggering glutamate secretion in the CNS and by potentiating depolarization-evoked glutamate secretion. Exogenously added S1P at a nanomolar concentration elicited glutamate secretion from hippocampal cells, and S1P at a picomolar level potentiated depolarization-evoked secretion in neurons [39]. These data suggest that variations in circulating or tissue levels of S1P could impact both thymic and CNS function, including T-ALL localization and infiltration (Fig. 1). The role of S1P/S1P1 in this context needs further investigation. Furthermore, S1P/S1P1 migratory responses of T-ALL primary cells (as well as T-LBL samples) can be controlled by SEMA3F/NRP2 [35], showing a more complex mechanism of regulation of S1P1 signaling. Since SEMAS are also present in both leukemia and CNS cells, one can imagine that they can mediate the crosstalk and impact organ invasion.

Fig. 1.

Sphingosine-1-phosphate (S1P) as a possible mediator of thymus-brain connections. Some molecular circuits, here represented by S1P and its receptor S1P1, may mediate the connections between the thymus and the brain in the case of T-ALL. S1P is present in the bloodstream, thymus, and brain in different concentrations, forming gradients to which cells respond. The migratory response of T-ALL lymphoblasts to S1P depends on the concentration of this lipid and the S1P1 receptor and may lead to chemotaxis or fugotaxis [38]. We hypothesize that S1P is involved in the exit of T-ALL blasts from the thymus and consequent leukemization. Variation in the circulating levels of S1P could also contribute to the infiltration of blasts in the CNS and to the alteration of the niche by neuroinflammation. The variation of S1P levels may also have indirect consequences such as the triggering of glutamate secretion in the CNS, and glutamate has a pro-metastatic effect, in addition to the possible effects on the thymus.

Fig. 1.

Sphingosine-1-phosphate (S1P) as a possible mediator of thymus-brain connections. Some molecular circuits, here represented by S1P and its receptor S1P1, may mediate the connections between the thymus and the brain in the case of T-ALL. S1P is present in the bloodstream, thymus, and brain in different concentrations, forming gradients to which cells respond. The migratory response of T-ALL lymphoblasts to S1P depends on the concentration of this lipid and the S1P1 receptor and may lead to chemotaxis or fugotaxis [38]. We hypothesize that S1P is involved in the exit of T-ALL blasts from the thymus and consequent leukemization. Variation in the circulating levels of S1P could also contribute to the infiltration of blasts in the CNS and to the alteration of the niche by neuroinflammation. The variation of S1P levels may also have indirect consequences such as the triggering of glutamate secretion in the CNS, and glutamate has a pro-metastatic effect, in addition to the possible effects on the thymus.

Close modal

Chemokines and Chemokine Receptors

Chemokines and chemokine receptors are constitutively expressed in the immune and nervous systems, being essential for the thymus and brain development and homeostasis. These proteins play a role in the migration, differentiation, and proliferation of thymocytes, TECs, glial, and neuronal cells [82, 83], and protection at CNS barriers [84]. They are also involved in cancer cell survival, proliferation, and migration [85], as well as in T-ALL development and migration [86].

Different studies have shown that blasts from patients with T-ALL express altered levels of CXCR4 [87‒89]. Similarly, the expression of CXCR4 was observed in T-ALL cell lines such as Jurkat, CEM, and MOLT-4 [43, 89]. The CXCR4-CXCL12 axis has been implicated as a potential pathway that drives T-ALL invasion into the CNS. Using a T-ALL in vivo model, where alymphoid NGC mice were injected with Jurkat cells expressing CXCR4 or deficient for this receptor, it was shown that the absence of CXCR4 increased survival and minimal signs of CNS pathology [44].

Another study by Jost et al. [43] showed that Jurkat and CEM cells injected into mice invaded the CNS and were located in the meninges. However, treatment with AMD3100, a CXCR4 antagonist, reduced the meninge infiltration [43]. Together, these data support the participation of the CXCR4/CXCL12 axis in targeting T-ALL cells in the CNS.

Most primary T-ALL samples and cell lines also overexpress CCR7, which is a Notch1-induced adhesion regulator. It is estimated that Notch1 signaling pathway activation occurs in at least 80% of all T-ALL cases, and a study has shown that CCR7 was expressed in 80% of T-ALL lines and 73% of primary T-ALL samples analyzed [47]. In this study, it was shown that transplantation of human T-ALL cells into alymphoid Rag2−/−Il2rg−/− mice led to distinct survival patterns depending on the CCR7 expression, where hosts that received CCR7+ cells succumbed to the disease earlier than those receiving CCR7 cells. CCR7 expression was sufficient for CNS infiltration in response to CCL19, detected mainly in brain venules near infiltrating lymphocytes. Interestingly, CCR7 function seems specific for Notch1-induced T-cell malignancies since deletion of this receptor in two models of B-ALL failed to suppress CNS infiltration.

CXCR4 and CCR7 expression levels in patient samples with CNS involvement were significantly increased compared to patients without CNS involvement. In addition, in NSG mice transplanted with Jurkat cells, leukemic cells infiltrated the bone marrow and the CNS. CCR7 blockade significantly reduced Jurkat infiltration into the CNS but not into the bone marrow, reinforcing the role of CCR7 in T-ALL infiltration into the CNS [45].

Thymic Hormones

Some endogenous components produced in the thymus, such as thymulin, thymosin, and thymopoietin hormones, play a role in thymocyte differentiation [90, 91]. Thymic hormones are also found in the CNS since thymulin can be produced by astrocytes and glial cells in the brain of rodents, and thymosin can be produced in different regions of the CNS, mainly in the hippocampus [48, 49]. Studies by Safieh-Garabedian et al. [92, 93] showed that pre-treatment with thymulin inhibited the induction of pro-inflammatory cytokines and reversed local inflammation in vivo in rat models using an intracerebroventricular (i.c.v.) injection of LPS or endotoxin. Moreover, early treatment with thymosin B4 was anti-inflammatory and reduced cortical lesions and loss of hippocampal cells in traumatic brain injury, suggesting a neuroprotective role of thymulin and thymosin in the CNS [94].

Significantly lower plasma levels of active thymulin have been observed in samples from patients with initial T-ALL or during relapse when compared to healthy controls, apparently due to a zinc deficiency [50]. Interestingly, active thymulin in physiologic doses (corresponding to the amount in the blood of young healthy individuals) reduced the proliferation of primary and T-ALL cell lines. Although there is evidence for the role of thymic hormones, such as thymulin, in T-ALL development and the CNS separately, further studies are necessary to explore the role of these hormones in the context of the thymus-brain connections in T-ALL.

Different aspects of the possible connections between the thymus (and T lymphoblasts) and the brain are explored herein, focusing on soluble molecules and their receptors. Among them, neurotransmitters, thymic hormones, chemokines, and other molecules shared between the thymus and the brain may participate in the crosstalk between these two organs and, consequently, in T-ALL development and invasion in the CNS.

Understanding the specific mechanisms and routes by which T-ALL cells infiltrate the CNS remains an important question and would be of great value to leukemia treatment. For example, understanding whether T-ALL cells reach the CNS by first invading the bone marrow or the CSF, and identifying the molecules involved in different migration patterns could help to identify biomarkers able to predict or detect CNS infiltration in T-ALL at an early stage, thereby helping to monitor disease progression. Understanding how leukemia cells interact with neural cells, immune cells, and other components of the CNS may also provide insights into disease progression and potential therapeutic targets. Therefore, a deeper understanding of the role of different mediators of thymus-brain connections could shed light on the mechanisms involved in the pathogenesis of the disease including lymphoblast dissemination and CNS infiltration, given their clinical importance.

The authors have no conflicts of interest to declare.

This work was supported by Fiocruz, Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) – Rio de Janeiro, and the Mercosur Fund for Structural Convergence (FOCEM). It was developed in the Brazilian National Institute of Science and Technology frameworks on neuroimmunomodulation (CNPq) and the Rio de Janeiro Research Network on Neuroinflammation (FAPERJ).

D.A.M.C. and V.C.A. contributed to the conception, organization, and writing of the manuscript. E.P.B. contributed to the conception and writing of the manuscript and organized references.

1.
International Agency for Research on Cancer
,
Travis
WD
,
Brambilla
E
,
Burke
A
,
Marx
A
,
Nicholson
AG
.
WHO classification of tumours of the lung
.
Pleura
:
Thymus and Heart
.
2.
Arber
DA
,
Orazi
A
,
Hasserjian
R
,
Thiele
J
,
Borowitz
MJ
,
Le Beau
MM
, et al
.
The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia
.
Blood
.
2016
;
127
(
20
):
2391
405
. .
3.
Jain
N
,
Lamb
AV
,
O’brien
S
,
Ravandi
F
,
Konopleva
M
,
Jabbour
E
, et al
.
Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: a high-risk subtype
.
Blood
.
2016
;
127
(
15
):
1863
9
. .
4.
Chen
J
,
Liu
L
,
Ma
R
,
Pang
A
,
Yang
D
,
Chen
X
, et al
.
Comparison of characteristics and outcomes on ETP-ALL/LBL and non-ETP ALL patients receiving allogeneic hematopoietic stem cell transplantation
.
Front Oncol
.
2022
;
12
:
1025885
. .
5.
Patrick
K
,
Wade
R
,
Goulden
N
,
Mitchell
C
,
Moorman
AV
,
Rowntree
C
, et al
.
Outcome for children and young people with Early T-cell precursor acute lymphoblastic leukaemia treated on a contemporary protocol, UKALL 2003
.
Br J Haematol
.
2014
;
166
(
3
):
421
4
. .
6.
Zuurbier
L
,
Gutierrez
A
,
Mullighan
CG
,
Canté-Barrett
K
,
Gevaert
AO
,
De Rooi
J
, et al
.
Immature MEF2C-dysregulated T-cell leukemia patients have an early T-cell precursor acute lymphoblastic leukemia gene signature and typically have non-rearranged t-cell receptors
.
Haematologica
.
2014
;
99
(
1
):
94
102
. .
7.
März
M
,
Meyer
S
,
Erb
U
,
Georgikou
C
,
Horstmann
MA
,
Hetjens
S
, et al
.
Pediatric acute lymphoblastic leukemia: conquering the CNS across the choroid plexus
.
Leuk Res
.
2018
;
71
:
47
54
. .
8.
Yao
H
,
Price
TT
,
Cantelli
G
,
Ngo
B
,
Warner
MJ
,
Olivere
L
, et al
.
Leukaemia hijacks a neural mechanism to invade the central nervous system
.
Nature
.
2018
;
560
(
7716
):
55
60
. .
9.
Rajakumar
SA
,
Grandal
I
,
Minden
MD
,
Hitzler
JK
,
Guidos
CJ
,
Danska
JS
.
Targeted blockade of immune mechanisms inhibit B precursor acute lymphoblastic leukemia cell invasion of the central nervous system
.
Cell Rep Med
.
2021
;
2
(
12
):
100470
. .
10.
Thastrup
M
,
Duguid
A
,
Mirian
C
,
Schmiegelow
K
,
Halsey
C
.
Central nervous system involvement in childhood acute lymphoblastic leukemia: challenges and solutions
.
Leukemia
.
2022
;
36
(
12
):
2751
68
. .
11.
Scharff
BFSS
,
Modvig
S
,
Marquart
HV
,
Christensen
C
.
Integrin-mediated adhesion and chemoresistance of acute lymphoblastic leukemia cells residing in the bone marrow or the central nervous system
.
Front Oncol
.
2020
;
10
:
775
10
. .
12.
Francelin
C
,
Veneziani
LP
,
Farias
A
,
Mendes-da-Cruz
DA
,
Savino
W
,
Savino
W
.
Neurotransmitters modulate intrathymic T-cell development
.
Front Cell Dev Biol
.
2021
;
9
:
668067
7
. .
13.
Hodo
TW
,
de Aquino
MTP
,
Shimamoto
A
,
Shanker
A
.
Critical neurotransmitters in the neuroimmune network
.
Front Immunol
.
2020
;
11
:
1869
. .
14.
Bosticardo
M
,
Notarangelo
LD
.
Human thymus in health and disease: recent advances in diagnosis and biology
.
Semin Immunol
.
2023
;
66
:
101732
. .
15.
Leposavić
G
,
Pilipović
I
,
Radojević
K
,
Pešić
V
,
Perišić
M
,
Kosec
D
.
Catecholamines as immunomodulators: a role for adrenoceptor-mediated mechanisms in fine tuning of T-cell development
.
Auton Neurosci
.
2008
;
144
(
1–2
):
1
12
. .
16.
Mignini
F
,
Sabbatini
M
,
Mattioli
L
,
Cosenza
M
,
Artico
M
,
Cavallotti
C
.
Neuro-immune modulation of the thymus microenvironment (Review)
.
Int J Mol Med
.
2014
;
33
(
6
):
1392
400
. .
17.
Godinho-Silva
C
,
Cardoso
F
,
Veiga-Fernandes
H
.
Neuro-immune cell units: a new paradigm in physiology
.
Annu Rev Immunol
.
2019
;
37
:
19
46
. .
18.
Al-Shalan
HAM
,
Hu
D
,
Nicholls
PK
,
Greene
WK
,
Ma
B
.
Immunofluorescent characterization of innervation and nerve-immune cell neighborhood in mouse thymus
.
Cell Tissue Res
.
2019
;
378
(
2
):
239
54
. .
19.
Tollefson
L
,
Bulloch
K
.
Dual label retrograde transport: CNS innervation of the mouse thymus distinct from other mediastinum viscera
.
J Neurosci Res
.
1990
;
25
(
1
):
20
8
. .
20.
Trotter
RN
,
Stornetta
RL
,
Guyenet
PG
,
Roberts
MR
.
Transneuronal mapping of the CNS network controlling sympathetic outflow to the rat thymus
.
Auton Neurosci
.
2007
;
131
(
1–2
):
9
20
. .
21.
Mićić
M
,
Leposavić
G
,
Ugresić
N
,
Bogojević
M
,
Isaković
K
.
Parasympathetic innervation of the rat thymus during first life period: histochemical and biochemical study
.
Thymus
.
1992
;
19
(
3
):
173
82
.
22.
Rinner
I
,
Globerson
A
,
Kawashima
K
,
Korsatko
W
,
Schauenstein
K
.
A possible role for acetylcholine in the dialogue between thymocytes and thymic stroma
.
Neuroimmunomodulation
.
1999
;
6
(
1–2
):
51
5
. .
23.
Kawashima
K
,
Fujii
T
.
Expression of non-neuronal acetylcholine in lymphocytes and its contribution to the regulation of immune function
.
Front Biosci
.
2004
;
9
:
2063
85
. .
24.
Panneck
AR
,
Rafiq
A
,
Schütz
B
,
Soultanova
A
,
Deckmann
K
,
Chubanov
V
, et al
.
Cholinergic epithelial cell with chemosensory traits in murine thymic medulla
.
Cell Tissue Res
.
2014
;
358
(
3
):
737
48
. .
25.
Xu
Y
,
Li
J
,
Luo
Y
,
Ma
J
,
Huang
P
,
Chen
Y
, et al
.
Carvedilol exhibits anti-acute T lymphoblastic leukemia effect in vitro and in vivo via inhibiting β-ARs signaling pathway
.
Biochem Biophys Res Commun
.
2023
;
639
:
150
60
. .
26.
Dobrovinskaya
O
,
Valencia-Cruz
G
,
Castro-Sánchez
L
,
Bonales-Alatorre
EO
,
Liñan-Rico
L
,
Pottosin
I
.
Cholinergic machinery as relevant target in acute lymphoblastic T leukemia
.
Front Pharmacol
.
2016
;
7
:
290
. .
27.
Fujii
T
,
Tsuchiya
T
,
Yamada
S
,
Fujimoto
K
,
Suzuki
T
,
Kasahara
T
, et al
.
Localization and synthesis of acetylcholine in human leukemic T cell lines
.
J Neurosci Res
.
1996
;
44
(
1
):
66
72
. .
28.
Kamimura
Y
,
Fujii
T
,
Kojima
H
,
Nagano
T
,
Kawashima
K
.
Nitric oxide (NO) synthase mRNA expression and NO production via muscarinic acetylcholine receptor-mediated pathways in the CEM, human leukemic T-cell line
.
Life Sci
.
2003
;
72
(
18–19
):
2151
4
. .
29.
Ganor
Y
,
Besser
M
,
Ben-Zakay
N
,
Unger
T
,
Levite
M
.
Human T cells express a functional ionotropic glutamate receptor GluR3, and glutamate by itself triggers integrin-mediated adhesion to laminin and fibronectin and chemotactic migration
.
J Immunol
.
2003
;
170
(
8
):
4362
72
. .
30.
Miglio
G
,
Dianzani
C
,
Fallarini
S
,
Fantozzi
R
,
Lombardi
G
.
Stimulation of N-methyl-d-aspartate receptors modulates Jurkat T cell growth and adhesion to fibronectin
.
Biochem Biophys Res Commun
.
2007
;
361
(
2
):
404
9
. .
31.
Santomasso
B
,
Park
J
,
Salloum
D
,
Riviere
I
,
Flynn
J
,
Mead
E
, et al
.
Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia
.
Cancer Discov
.
2018
;
8
(
8
):
958
71
. .
32.
Levite
M
,
Chowers
Y
,
Ganor
Y
,
Besser
M
,
Hershkovits
R
,
Cahalon
L
.
Dopamine interacts directly with its D3 and D2 receptors on normal human T cells, and activates beta1 integrin function
.
Eur J Immunol
.
2001
;
31
:
3504
12
. .
33.
Basu
B
,
Sarkar
C
,
Chakroborty
D
,
Ganguly
S
,
Shome
S
,
Dasgupta
PS
, et al
.
D1 and D2 dopamine receptor-mediated inhibition of activated normal T cell proliferation is lost in Jurkat T leukemic cells
.
J Biol Chem
.
2010
;
285
(
35
):
27026
32
. .
34.
Cheung
YT
,
Khan
RB
,
Liu
W
,
Brinkman
TM
,
Edelmann
MN
,
Reddick
WE
, et al
.
Association of cerebrospinal fluid biomarkers of central nervous system injury with neurocognitive and brain imaging outcomes in children receiving chemotherapy for acute lymphoblastic leukemia
.
JAMA Oncol
.
2018
;
4
(
7
):
e180089
. .
35.
Mendes-da-Cruz
DA
,
Brignier
AC
,
Asnafi
V
,
Baleydier
F
,
Messias
CV
,
Lepelletier
Y
, et al
.
Semaphorin 3F and neuropilin-2 control the migration of human T-cell precursors
.
PLoS One
.
2014
;
9
:
e103405
7
. .
36.
Karjalainen
K
,
Jaalouk
DE
,
Bueso-Ramos
CE
,
Zurita
AJ
,
Kuniyasu
A
,
Eckhardt
BL
, et al
.
Targeting neuropilin-1 in human leukemia and lymphoma
.
Blood
.
2011
;
117
(
3
):
920
7
. .
37.
Taniguchi
M
,
Yuasa
S
,
Fujisawa
H
,
Naruse
I
,
Saga
S
,
Mishina
M
, et al
.
Disruption of semaphorin III/D gene causes severe abnormality in peripheral nerve projection
.
Neuron
.
1997
;
19
(
3
):
519
30
. .
38.
Messias
CV
,
Santana-Van-Vliet
E
,
Lemos
JP
,
Moreira
OC
,
Cotta-De-Almeida
V
,
Savino
W
, et al
.
Sphingosine-1-phosphate induces dose-dependent chemotaxis or fugetaxis of T-ALL blasts through S1P1 activation
.
PLoS One
.
2016
;
11
(
1
):
e0148137
. .
39.
Kajimoto
T
,
Okada
T
,
Yu
H
,
Goparaju
SK
,
Jahangeer
S
,
Nakamura
S
.
Involvement of sphingosine-1-phosphate in glutamate secretion in hippocampal neurons
.
Mol Cell Biol
.
2007
;
27
(
9
):
3429
40
. .
40.
Chan
J
,
Hu
Z
,
Sieburth
D
.
Recruitment of sphingosine kinase to presynaptic terminals by a conserved muscarinic signaling pathway promotes neurotransmitter release
.
Genes Dev
.
2012
;
26
(
10
):
1070
85
. .
41.
Groves
A
,
Kihara
Y
,
Chun
J
.
Fingolimod: direct CNS effects of sphingosine 1-phosphate (S1P) receptor modulation and implications in multiple sclerosis therapy
.
J Neurol Sci
.
2013
;
328
(
1–2
):
9
18
. .
42.
Skoug
C
,
Martinsson
I
,
Gouras
G
,
Meissner
A
,
Duarte
J
.
Sphingosine 1-phoshpate receptors are located in synapses and control spontaneous activity of mouse neurons in culture
.
Neurochem Res
.
2022
;
47
(
10
):
3114
25
. .
43.
Jost
TR
,
Borga
C
,
Radaelli
E
,
Romagnani
A
,
Perruzza
L
,
Omodho
L
, et al
.
Role of CXCR4-mediated bone marrow colonization in CNS infiltration by T cell acute lymphoblastic leukemia
.
J Leukoc Biol
.
2016
;
99
(
6
):
1077
87
. .
44.
Walker
KL
,
Rinella
SP
,
Hess
NJ
,
Turicek
DP
,
Kabakov
SA
,
Zhu
F
, et al
.
CXCR4 allows T cell acute lymphoblastic leukemia to escape from JAK1/2 and BCL2 inhibition through CNS infiltration
.
Leuk Lymphoma
.
2021
;
62
(
5
):
1167
77
. .
45.
Alsadeq
A
,
Schewe
DM
.
Acute lymphoblastic leukemia of the central nervous system: on the role of PBX1
.
Haematologica
.
2017
;
102
(
4
):
611
3
. .
46.
Passaro
D
,
Irigoyen
M
,
Catherinet
C
,
Gachet
S
,
Da Costa De Jesus
C
,
Lasgi
C
, et al
.
CXCR4 is required for leukemia-initiating cell activity in T cell acute lymphoblastic leukemia
.
Cancer Cell
.
2015
;
27
(
6
):
769
79
. .
47.
Buonamici
S
,
Trimarchi
T
,
Ruocco
MG
,
Reavie
L
,
Cathelin
S
,
Mar
BG
, et al
.
CCR7 signalling as an essential regulator of CNS infiltration in T-cell leukaemia
.
Nature
.
2009
;
459
(
7249
):
1000
4
. .
48.
Dardenne
M
,
Saade
N
,
Safieh-Garabedian
B
.
Role of thymulin or its analogue as a new analgesic molecule
.
Ann N Y Acad Sci
.
2006
;
1088
:
153
63
. .
49.
Turrini
P
,
Aloe
L
.
Evidence that endogenous thymosin alpha-1 is present in the rat central nervous system
.
Neurochem Int
.
1999
;
35
(
6
):
463
70
. .
50.
Mocchegiani
E
,
Paolucci
P
,
Granchi
D
,
Cavallazzi
L
,
Santarelli
L
,
Fabris
N
.
Plasma zinc level and thymic hormone activity in young cancer patients
.
Blood
.
1994
;
83
(
3
):
749
57
. .
51.
Elenkov
IJ
,
Wilder
RL
,
Chrousos
GP
,
Vizi
ES
.
The sympathetic nerve: an integrative interface between two supersystems: the brain and the immune system
.
Pharmacol Rev
.
2000
;
52
(
4
):
595
638
.
52.
Kocak
MZ
,
Er
M
,
Ugrakli
M
,
Hendem
E
,
Araz
M
,
Eryilmaz
MK
, et al
.
Could the concomitant use of beta blockers with bevacizumab improve survival in metastatic colon cancer
.
Eur J Clin Pharmacol
.
2023
;
79
(
4
):
485
91
. .
53.
Bustamante
P
,
Miyamoto
D
,
Goyeneche
A
,
de Alba Graue
PG
,
Jin
E
,
Tsering
T
, et al
.
Beta-blockers exert potent anti-tumor effects in cutaneous and uveal melanoma
.
Cancer Med
.
2019
;
8
(
17
):
7265
77
. .
54.
Koh
M
,
Takahashi
T
,
Kurokawa
Y
,
Kobayashi
T
,
Saito
T
,
Ishida
T
, et al
.
Propranolol suppresses gastric cancer cell growth by regulating proliferation and apoptosis
.
Gastric Cancer
.
2021
;
24
(
5
):
1037
49
. .
55.
Pérez Piñero
C
,
Rivero
EM
,
Gargiulo
L
,
Rodríguez
MS
,
Bruque
CD
,
Bruzzone
A
, et al
.
Adrenergic receptors in breast cancer
.
Prog Mol Biol Transl Sci
.
2022
;
193
(
1
):
37
63
. .
56.
Hajighasemi
F
,
Hajighasemi
S
.
Effect of propranolol on angiogenic factors in human hematopoietic cell lines in vitro
.
Iran Biomed J
.
2009
;
13
(
4
):
223
8
.
57.
Hajighasemi
F
,
Mirshafiey
A
.
In vitro sensitivity of leukemia cells to propranolol
.
J Clin Med Res
.
2009
;
1
(
3
):
144
9
. .
58.
Song
G
,
Li
Y
,
Jiang
G
.
Role of VEGF/VEGFR in the pathogenesis of leukemias and as treatment targets ( Review)
.
Oncol Rep
.
2012
;
28
(
6
):
1935
44
. .
59.
Münch
V
,
Trentin
L
,
Herzig
J
,
Demir
S
,
Seyfried
F
,
Kraus
JM
, et al
.
Central nervous system involvement in acute lymphoblastic leukemia is mediated by vascular endothelial growth factor
.
Blood
.
2017
;
130
(
5
):
643
54
. .
60.
Siddhartha
R
,
Garg
M
.
Molecular and clinical insights of matrix metalloproteinases into cancer spread and potential therapeutic interventions
.
Toxicol Appl Pharmacol
.
2021
;
426
:
115593
. .
61.
Kawashima
K
,
Fujii
T
.
Extraneuronal cholinergic system in lymphocytes
.
Pharmacol Ther
.
2000
;
86
(
1
):
29
48
. .
62.
Battisti
V
,
Schetinger
MRC
,
Maders
LDK
,
Santos
KF
,
Bagatini
MD
,
Correa
MC
, et al
.
Changes in acetylcholinesterase (AchE) activity in lymphocytes and whole blood in acute lymphoblastic leukemia patients
.
Clin Chim Acta
.
2009
;
402
(
1–2
):
114
8
. .
63.
Peng
H
,
Ferris
RL
,
Matthews
T
,
Hiel
H
,
Lopez-Albaitero
A
,
Lustig
LR
.
Characterization of the human nicotinic acetylcholine receptor subunit alpha (alpha) 9 (CHRNA9) and alpha (alpha) 10 (CHRNA10) in lymphocytes
.
Life Sci
.
2004
;
76
(
3
):
263
80
. .
64.
Landais
E
,
Liautaud-Roger
F
,
Frank
A
.
Lymphocytes prime activation is required for nicotine-induced calcium waves
.
Front Biosci
.
2010
;
E2
:
152
. .
65.
Alea
MP
,
Borroto-Escuela
DO
,
Romero-Fernandez
W
,
Fuxe
K
,
Garriga
P
.
Differential expression of muscarinic acetylcholine receptor subtypes in Jurkat cells and their signaling
.
J Neuroimmunol
.
2011
;
237
(
1–2
):
13
22
. .
66.
Suriyo
T
,
Chotirat
S
,
Auewarakul
CU
,
Chaiyot
K
,
Promsuwicha
O
,
Satayavivad
J
.
Variation of nicotinic subtype α7 and muscarinic subtype M3 acetylcholine receptor expression in three main types of leukemia
.
Oncol Lett
.
2019
;
17
(
1
):
1357
62
. .
67.
Tarnawski
L
,
Shavva
VS
,
Kort
EJ
,
Zhuge
Z
,
Nilsson
I
,
Gallina
AL
, et al
.
Cholinergic regulation of vascular endothelial function by human ChAT+ T cells
.
Proc Natl Acad Sci U S A
.
2023
;
120
(
14
):
e2212476120
. .
68.
Ganor
Y
,
Levite
M
.
The neurotransmitter glutamate and human T cells: glutamate receptors and glutamate-induced direct and potent effects on normal human T cells, cancerous human leukemia and lymphoma T cells, and autoimmune human T cells
.
J Neural Transm
.
2014
;
121
(
8
):
983
1006
. .
69.
Ganor
Y
,
Grinberg
I
,
Reis
A
,
Grinberg
I
,
Cooper
I
,
Levite
M
.
Human T-leukemia and T-lymphoma express glutamate receptor AMPA GluR3, and the neurotransmitter glutamate elevates the cancer-related matrix-metalloproteinases inducer CD147/EMMPRIN, MMP-9 secretion and engraftment of T-leukemia in vivo
.
Leuk Lymphoma
.
2009
;
50
(
6
):
985
97
. .
70.
Zheng
NS
,
Zhao
XY
,
Wei
D
,
Miao
JL
,
Liu
ZK
,
Yong
YL
, et al
.
CD147-specific chimeric antigen receptor T cells effectively inhibit T cell acute lymphoblastic leukemia
.
Cancer Lett
.
2022
;
542
:
215762
. .
71.
Grant
CE
,
Flis
AL
,
Ryan
BM
.
Understanding the role of dopamine in cancer: past, present and future
.
Carcinogenesis
.
2022
;
43
(
6
):
517
27
. .
72.
Li
M
,
Zhou
L
,
Sun
X
,
Yang
Y
,
Zhang
C
,
Wang
T
, et al
.
Dopamine, a co-regulatory component, bridges the central nervous system and the immune system
.
Biomed Pharmacother
.
2022
;
145
:
112458
. .
73.
Klein
MO
,
Battagello
DS
,
Cardoso
AR
,
Hauser
DN
,
Bittencourt
JC
,
Correa
RG
.
Dopamine: functions, signaling, and association with neurological diseases
.
Cell Mol Neurobiol
.
2019
;
39
(
1
):
31
59
. .
74.
Franco
R
,
Reyes-Resina
I
,
Navarro
G
.
Dopamine in health and disease: much more than a neurotransmitter
.
Biomedicines
.
2021
;
9
(
2
):
109
13
. .
75.
Besser
MJ
,
Ganor
Y
,
Levite
M
.
Dopamine by itself activates either D2, D3 or D1/D5 dopaminergic receptors in normal human T-cells and triggers the selective secretion of either IL-10, TNFalpha or both
.
J Neuroimmunol
.
2005
;
169
(
1–2
):
161
71
. .
76.
Watanabe
Y
,
Nakayama
T
,
Nagakubo
D
,
Hieshima
K
,
Jin
Z
,
Katou
F
, et al
.
Dopamine selectively induces migration and homing of naive CD8+ T cells via dopamine receptor D3
.
J Immunol
.
2006
;
176
(
2
):
848
56
. .
77.
Lifantseva
NV
,
Koneeva
TO
,
Voronova
SN
,
Zakharova
LA
,
Melnikova
VI
.
The inhibition of dopamine synthesis in fetuses changes the pattern of T-lymphocyte maturation in the thymus of adult rats
.
Dokl Biochem Biophys
.
2016
;
470
(
1
):
342
4
. .
78.
Mendes-da-Cruz
DA
,
Linhares-Lacerda
L
,
Smaniotto
S
,
Dardenne
M
,
Savino
W
.
Semaphorins and neuropilins: new players in the neuroendocrine control of the intrathymic T-cell migration in humans
.
Exp Physiol
.
2012
;
97
(
11
):
1146
50
. .
79.
Lambrechts
D
,
Carmeliet
P
.
VEGF at the neurovascular interface: therapeutic implications for motor neuron disease
.
Biochim Biophys Acta
.
2006
;
1762
(
11–12
):
1109
21
. .
80.
Tang
Y
,
Jiang
F
,
Guo
L
,
Si
M
,
Jiao
X
.
Expression and significance of vascular endothelial growth factor A and C in leukemia central nervous system metastasis
.
Leuk Res
.
2013
;
37
(
4
):
359
66
. .
81.
Feng
H
,
Stachura
DL
,
White
RM
,
Gutierrez
A
,
Zhang
L
,
Sanda
T
, et al
.
T-lymphoblastic lymphoma cells express high levels of BCL2, S1P1, and ICAM1, leading to a blockade of tumor cell intravasation
.
Cancer Cell
.
2010
;
18
(
4
):
353
66
. .
82.
Lalor
SJ
,
Segal
BM
.
Lymphoid chemokines in the CNS
.
J Neuroimmunol
.
2010
;
224
(
1–2
):
56
61
. .
83.
Savino
W
,
Mendes-Da-Cruz
DA
,
Silva
JS
,
Dardenne
M
,
Cotta-De-Almeida
V
.
Intrathymic T-cell migration: a combinatorial interplay of extracellular matrix and chemokines
.
Trends Immunol
.
2002
;
23
(
6
):
305
13
. .
84.
Williams
JL
,
Holman
DW
,
Klein
RS
.
Chemokines in the balance: maintenance of homeostasis and protection at CNS barriers
.
Front Cell Neurosci
.
2014
;
8
:
154
12
. .
85.
Marcuzzi
E
,
Angioni
R
,
Molon
B
,
Calì
B
.
Chemokines and chemokine receptors: orchestrating tumor metastasization
.
Int J Mol Sci
.
2018
;
20
(
1
):
96
. .
86.
Harasawa
H
,
Yamada
Y
,
Hieshima
K
,
Jin
ZHE
,
Nakayama
T
,
Yoshie
O
, et al
.
Survey of chemokine receptor expression reveals frequent co-expression of skin-homing CCR4 and CCR10 in adult T-cell leukemia/lymphoma
.
Leuk Lymphoma
.
2006
;
47
(
10
):
2163
73
. .
87.
Crazzolara
R
,
Kreczy
A
,
Mann
G
,
Heitger
A
,
Eibl
G
,
Fink
FM
, et al
.
High expression of the chemokine receptor CXCR4 predicts extramedullary organ infiltration in childhood acute lymphoblastic leukaemia
.
Br J Haematol
.
2001
;
115
(
3
):
545
53
. .
88.
Dialynas
DP
,
Shao
L
,
Billman
GF
,
Yu
J
.
Engraftment of human T-cell acute lymphoblastic leukemia in immunodeficient NOD/SCID mice which have been preconditioned by injection of human cord blood
.
Stem Cell
.
2001
;
19
(
5
):
443
52
. .
89.
Melo
RDCC
,
Longhini
AL
,
Bigarella
CL
,
Baratti
MO
,
Traina
F
,
Favaro
P
, et al
.
CXCR7 is highly expressed in acute lymphoblastic leukemia and potentiates CXCR4 response to CXCL12
.
PLoS One
.
2014
;
9
(
1
):
e85926
. .
90.
Savino
W
,
Dardenne
M
.
Neuroendocrine control of thymus physiology
.
Endocr Rev
.
2000
;
21
(
4
):
412
43
. .
91.
Rezzani
R
,
Franco
C
,
Hardeland
R
,
Rodella
LF
.
Thymus-pineal gland axis: revisiting its role in human life and ageing
.
Int J Mol Sci
.
2020
;
21
(
22
):
8806
40
. .
92.
Safieh-Garabedian
B
,
Jabbur
SJ
,
Dardenne
M
,
Saadé
NE
.
Thymulin related peptide attenuates inflammation in the brain induced by intracerebroventricular endotoxin injection
.
Neuropharmacology
.
2011
;
60
(
2–3
):
496
504
. .
93.
Safieh-Garabedian
B
,
Ochoa-Chaar
CI
,
Poole
S
,
Massaad
CA
,
Atweh
SF
,
Jabbur
SJ
, et al
.
Thymulin reverses inflammatory hyperalgesia and modulates the increased concentration of proinflammatory cytokines induced by i.c.v. endotoxin injection
.
Neuroscience
.
2003
;
121
(
4
):
865
73
. .
94.
Xiong
Y
,
Mahmood
A
,
Meng
Y
,
Zhang
Y
,
Zhang
ZG
,
Morris
DC
, et al
.
Neuroprotective and neurorestorative effects of thymosin β4 treatment following experimental traumatic brain injury
.
Ann N Y Acad Sci
.
2012
;
1270
:
51
8
. .