Skip to main content

REVIEW article

Front. Immunol., 05 August 2024
Sec. Systems Immunology

Exploring the bone marrow micro environment in thalassemia patients: potential therapeutic alternatives

Zengzheng Li,,Zengzheng Li1,2,3Xiangmei Yao,,Xiangmei Yao1,2,3Jie ZhangJie Zhang4Jinghui YangJinghui Yang5Junxue NiJunxue Ni6Yajie Wang,,*Yajie Wang1,2,3*
  • 1Department of Hematology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
  • 2Yunnan Province Clinical Research Center for Hematologic Disease, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
  • 3Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China
  • 4Department of Medical Genetics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
  • 5Department of Pediatrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
  • 6Hospital Office, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China

Genetic mutations in the β-globin gene lead to a decrease or removal of the β-globin chain, causing the build-up of unstable alpha-hemoglobin. This condition is referred to as beta-thalassemia (BT). The present treatment strategies primarily target the correction of defective erythropoiesis, with a particular emphasis on gene therapy and hematopoietic stem cell transplantation. However, the presence of inefficient erythropoiesis in BT bone marrow (BM) is likely to disturb the previously functioning BM microenvironment. This includes accumulation of various macromolecules, damage to hematopoietic function, destruction of bone cell production and damage to osteoblast(OBs), and so on. In addition, the changes of BT BM microenvironment may have a certain correlation with the occurrence of hematological malignancies. Correction of the microenvironment can be achieved through treatments such as iron chelation, antioxidants, hypoglycemia, and biologics. Hence, This review describes damage in the BT BM microenvironment and some potential remedies.

1 Introduction

BT is an autosomal recessive hematological condition characterized by an imbalance between α-globin and β-globin chains, leading to inefficient erythropoiesis (1, 2). BT can cause iron overload (IO), extramedullary hematopoiesis, BM expansion, hemolytic anemia, and multiple organ involvement, which has a significant clinical impact on patients (24). In BT patients, the unbound α-globin binds to free heme molecules to form toxic insoluble aggregates (called hemipigment), which precipitate and destroy the red blood cell (RBC) membrane. Simultaneously, they also initiate the production of reactive oxygen species (ROS), leading to oxidative stress and impacting the longevity of specific subsets of RBC (5, 6). Heat shock protein 70 (HSP70) is consistently expressed in human erythroblasts. As these erythroblasts mature, HSP70 moves into the nucleus and protects GATA-1, the primary transcription factor responsible for erythrocyte production, by preventing caspase-3 cleavage (7). An excess of free α-globin chains binds to HSP70, leaving GATA-1 unprotected which in turn causes cleavage and degradation by caspase-3. Ultimately, end-stage maturation arrest and erythroid progenitor cell apoptosis further impair RBC production (7). Additionally, ROS facilitates the excessive production of growth differentiation factor 11 (GDF11) in BT. This excessive production of GDF11 hinders the process of erythropoiesis in BT by activating SMAD2/3 signal transduction, which is involved in controlling the differentiation of RBC. As a result, the differentiation of erythrocytes is restricted (8, 9). It also promoted erythroid amplification and ineffective erythrocyte production in BT (8). This series of events leads to early apoptosis of mature nucleated erythrocytes, accompanied by hematopoietic amplification, followed by chronic hemolytic anemia with significant reticulocytosis, severe anemia, and a series of secondary pathophysiological mechanisms (10).

Heterogeneous populations of stromal cells and extracellular matrix form a specific microenvironment in the BM (11). The hematopoietic stem cells found in the BM establish a dynamic relationship with the surrounding microenvironment, ensuring the equilibrium of the body’s hematopoietic system (12). However, the BM microenvironment changes under some pathological conditions (13). For example, the tumor cells of acute myeloid leukemia are derived from the malignant transformation of hematopoietic stem cells and are able to alter the microenvironment, allowing the BM microenvironment to develop into a more suitable tumor microenvironment for tumor cell growth (14). This provides a refuge for malignant cells, allowing them to enter a chemotherapy-resistant state and become more prone to recurrence (15, 16). Unlike this, BT is a change in the BM microenvironment caused by ineffective erythrocyte production (17). This may not provide benefits for ineffective erythropoiesis, but rather consistently damage the microenvironment. This review focuses on the possible correlations between the accumulation of macromolecules in the BM, impaired maintenance of hematopoietic function, disruption of bone cell differentiation, destruction of bone structure, thalassemia, and hematologic tumors. Furthermore, possible approaches that might potentially provide therapeutic benefits are also discussed. This may aid in comprehending and managing the BM microenvironment of BT.

2 Bone marrow microevent and cellular composition and lineage in normal condition

Cell proliferation and BM activity are increased in the BM of patients with BT during inefficient erythropoiesis, despite the fact that the process is not functional, it is not completely halted (18). Attenuated Total Reflection-Fourier Transform Infrared Spectroscopy (ATR-FTIR) found that lipid, protein, glycogen, and nucleic acid contents in thalassemia BM mesenchymal stem cells (MSCs) were significantly higher than those in normal MSCs (19). The content of these macromolecules in BM MSCs of BT patients after hematopoietic stem cell transplantation was significantly lower than that before transplantation (19). This indicate that hematopoietic stem cell transplantation can effectively address the accumulation of macromolecules in MSCs. Furthermore, the degree of fat unsaturation in the BM increases proportionally with the increase in erythropoiesis (20). In patients with BT, the amount of fat and apolipoprotein D (APOD) in the BM decreases, but the levels of unsaturated fatty acids(UFA) increase. This phenomenon is associated with ineffective erythropoiesis (21, 22). Studies have shown that hydroxyurea therapy can improve the abnormal metabolic pathways of lipoprotein changes, glycolysis, Tricarboxylic acid cycle, fatty acid, and choline metabolism in BT patients (2325). Additionally, individuals with transfusion-dependent thalassemia (TDT) commonly experience diabetic mellitus (DM) as a consequence, and there is a significant accumulation of glycogen in the BM microenvironment (19, 26). Patients with thalassemia need to be provided with appropriate treatment and monitored for a long time. Some patients develop diabetes even after hematopoietic stem cell transplantation (27). Oral hypoglycemic agents are effective and safe in the treatment of DM in TDT patients and can achieve adequate blood glucose control in a considerable time (28). Due to the accumulation of glycogen in the BM microenvironment, long-term blood glucose monitoring and appropriate treatment should be given to patients with thalassemia. Metformin is an oral hypoglycemic drug with multiple effects. Additionally, it can help preserve the integrity of DNA (29). It has a beneficial impact on cardiac function and reduces the chances of heart failure and renal damage (30, 31). Engliflozin (Em) also has the effect of controlling blood sugar and can reverse the PlGF-1 resistance phenotype of hyperglycemic monocytes. Moreover, Em also can restore EC dysfunction in hyperglycemia, which may be attributed to the recovery of VEGFR-2 receptors on the EC surface (32). Most SGLT-1 and 2 inhibitors, including Sotagliflozin, have good effects on improving ROS, hyperglycemia, EC dysfunction, and heart failure (33, 34).

In the BM of BT patients, excessive α-globin chain accumulates in the progenitor RBC, resulting in the premature death of the progenitor RBC in the middle and late stage of BM, further aggravating the increase of the levels of erythropoietin (EPO) and growth differentiation factor 15 (GDF-15) (35). Furthermore, the levels of ferritin in the BM plasma of patients with BT were markedly elevated compared to both normal people and BT patients who had undergone transplantation, regardless of their usage of iron chelators (3638). At the same time, the content of ferritin in the BM plasma of BT was also significantly higher than that in peripheral blood (39). This evidence indicates that there is a large amount of iron accumulation in the BM of BT patients. An important explanation for this outcome is that MSCs can take up iron through ferroportin (For example SLC40A1, a protective mechanism against accumulation of cytoplasmic free iron) and transferrin receptor 1 (TFR1), and express the ferritin gene to store iron (40). This may be a protective measure against IO. However, prolonged exposure to iron can change MSCs’ systems for sensing and storing iron, which is primarily shown by their failure to stimulate SLC40A1 expression. This can lead to a buildup of cytoplasmic free iron (40, 41). Increased ROS levels will accompany iron accumulation in transfusion-independent patients, and oxidative damage from ROS is a major factor in thalassemia patients’ cell and tissue damage (42). In recent studies, it has been observed that the production of ROS can also occur through the action of cytochrome P450 (CYP450) 4A and 4F, which induce the production of 20-hydroxyeicosatetraenoic acid (20-HETE) (43). This overproduction of ROS was observed in Hbbth3/+ mice “model of β-thalassemia” and was found to be mediated by a pathway that is dependent on nicotinamide adenine dinucleotide phosphate (NADPH) (43, 44). Further increases in ROS levels in BT-MSCs resulted in decreased expression of antioxidant genes, altered ferroportin activation, and inappropriate regulation of iron-related genes such as TFR1 in BT-MSCs (42, 45, 46). The iron-overloaded BM environment impairs iron-sensing mechanisms, which may generate oxidative stress and alter the functional properties of MSCs (47, 48).

The accumulation of iron poses great harm to the BM microenvironment, leading to a series of deleterious effects including ROS. Iron chelation therapy is advantageous for ameliorating impaired lung function, renal function, cardiac function, vascular function, endocrine function, ROS, and ROS-induced chain reactions resulting from IO (4951). At present, the most frequently used drugs for iron chelation in thalassemia are deferoxamine (DFO), deferiprone (DFP), and deferasirox (DFX), which can reduce the incidence rate and mortality related to organ iron deposition, including BT patients with Hematopoietic stem cell transplantation (52). It has been seen that DFX has good safety and controllability when used in therapy (5355). Additionally, the approved luspatercept is an erythroid maturation agent that can be combined with selected transforming growth factor beta (TGF-β) superfamily ligands to reduce Smad 2/3 signaling and enhance late-stage erythropoiesis. Adult patients diagnosed with BT major (β-TM) now have a novel therapy available for long-term management (56). This medication seeks to reduce the necessity for frequent RBC transfusions, decrease anemia, and avoid excessive iron accumulation (56). Ferritin agonists can not only improve IO but also manage intermediate β-Anemia and liver burden in patients with thalassemia (57).

3 Bone micro environment in thalassemia patient

3.1 Hematopoietic stem cell quiescent state loss

Compared with normal mice, hematopoietic stem cells(HSCs) from Hbbth3/+ mice lose their quiescent state and enter the cell cycle. The primary observation is that the frequency of HSCs in the G0/G1 phase is significantly diminished, while the proportion of cells in the S phase increases and the number of colony-forming units is reduced (17, 58). Transplantation of HSCs derived from Hbbth3/+ mice into normal mice restored the long-term repopulation capacity of HSCs, whereas HSCs from normal mice transplanted into Hbbth3/+ mice inhibited the reconstitution process (17). These pieces of evidence are adequate to indicate that the function of HSCs is compromised in the myeloid environment of BT. This design provides more evidence that the BM microenvironment of the BT has an impact on the biological activity of HSCs. Due to chronically active ROS, patients with transfusion-dependent BT require higher stimulation of the CD34+ response to stress and have higher circulating rates of primitive hematopoietic stem progenitor cells (HSPCs) (39). Single-cell sequencing approaches revealed a higher percentage of CD34+ B lymphoid progenitors and a lower percentage of other stem and progenitor cell types in the CD34 compartment in pediatric BT patients (59). Studies have shown that daily doses of recombinant parathyroid hormone (PTH) can rescue HSCs defects by restoring the expression of stem cell genes (17). The main reason was that after Hbbth3/+ mice received PTH treatment, the number of resting HSCs increased and the level of cyclin-dependent protein kinase inhibitor 1C (CDKN1C) was also recovered (17, 60).

MSCs have been used in vitro to support the expansion of HSCs and HSPCs, and in vivo to promote HSPCs implantation (61). BM-MSCs are also damaged in BT patients, so will HSCs transplantation with BM-MSCs improve transplant outcomes? Clinical studies have found that the use of combined BM-derived MSCs does not affect the transplant outcome of type III thalassemia (62, 63). However, it has been found that bioactive molecules in extracellular vesicles derived from MSCS can regulate the expression of HSCs genes BIRC34, BIRC2, and NF-κB to improve the cloning ability of CD34+ cells (6466). However, the practical use of exosomes for treatment has not yet achieved any significant advancements.

3.2 Impaired MSCs function

The preservation of hematopoietic function is closely linked to the biological role of MSCs. Recent research has revealed a decrease in the occurrence of CD146+ and CD271+ cells in the BM of BT patients, and this decrease is inversely associated with levels of ROS (17). Moreover, the expression frequencies of CD73 and Sca-1 in MSCs of BM of Hbbth3/+ mice were also found to be decreased (17). The reduced levels of KIT ligand (KITLG) and CXC chemokine ligand 12 (CXCL12), which are crucial molecules for the maintenance of hematopoiesis, hindered the implantation, retention, survival, and proliferation of HSCs, are also observed (39, 67). Angiopoietin-1 (ANGPT1) and vascular endothelial growth factor (VEGFA), which regulate the quiescence of HSPC, were inhibited as well (39, 68). Fibroblast growth factor 2 (FGF2) and interleukin 6 (IL-6), which have amplification effects on HSPC and paracrine proliferation effects on MSC, are also inhibited (39, 69). Additionally, the amount of CD34+ attracted by BT MSCs is much lower compared to normal BM-MSCs, and its effect in promoting the expansion of umbilical cord blood HSCs is also poor (17, 39, 70). Inhibition of these molecules may be one of the reasons for the poor efficacy of BT HSCs transplantation.

The ability of CD105+ MSCs obtained from the BM of BT patients to differentiate into osteogenic cells is significantly limited (71). Runt-related transcription factor 2 (RUNX2), which is efficiently activated in normal MSCs, is a key factor regulating osteocyte production (72, 73). The expression of RUNX2 in MSCs was seen to be decreased in BT, leading to a downregulation of the secreted protein acidic and rich in cysteine (SPARC) and collagen type I alpha 2 (COL1A2) (39). Consequently, the formation of mineralized bone is hindered (39). The subchondral trabecular network density, bone mineral density, and trabecular number were decreased in the BM of Hbbth3/+ mice while the distance between trabeculae is enlarged (17, 74). This indicates that the process of MSCs transforming into OBs in the BM environment of patients with BT is impeded. Compared with the normal BM microenvironment, the expression of Notch ligand Jagged 1 is reduced in the BT mouse microenvironment (17). Loss of Jagged1 favors the induction of OBs ablation (17, 75). In a study, the formation of MSC-derived bone structures of BT in a humanized ossicle mouse model was significantly delayed (39, 76). The main manifestations were reduced bone-cell formation, hollow bone cavity, reduced number of blood vessels, and the formation of immature bone and abundant extracellular matrix. Moreover, few hematopoietic cells colonized, consistent with the impaired maintenance of hematopoiesis described above (39). The methylation of histone 3 lysine 9 (H3K9) and histone 3 lysine 36 (H3K36) was significantly downregulated in iron-overloaded BT-BM MSCs. This suggests an altered ability of MSCs to form appropriate niches in vivo.

3.3 Impaired OBs formation

The first unsuccessful erythropoiesis in thalassemia patients results in BM enlargement, which lowers trabecular bone tissue and thins the cortical layer. Second, endocrine disruption brought on by high iron loading results in higher bone turnover (77). MSCs derived from the BM of BT patients have weakened cloning ability, low proliferation rate, and inefficient differentiation ability, so the MSCs in the BM of BT cannot effectively differentiate into OBs in vivo. Moreover, the same bones are formed as the bone disease in BT patients (39). Further development includes osteoporosis, growth failure, spinal deformity, and fragility fracture diseases (78, 79). This is a significant factor contributing to the prevalence of bone-related diseases in BT patients (78, 79). Reduced OBs activity is the primary cause of low bone mineral density, and it may not be associated with osteoclast (OC) activity (17). The levels of alkaline phosphatase (ALP), an indicator of OBs activity, and the bone matrix glycoprotein osteopontin(OPN), mostly synthesized by OBs, were decreased in the Hbbth3/+ mice. Additionally, the absence of OPN resulted in HSCs progression into the cell cycle (80). Studies have shown that PTH can rescue HSCs function by increasing OPN levels (60). In thalassemia patients, the primary impairment of osteocyte lineage cells is OBs.

OBs themselves express transferrin receptor (TFR) and divalent metal transporter 1 (DMT1) (81). In the iron-overloaded BM microenvironment, iron toxicity can affect OBs to undergo apoptosis by directly altering bone microarchitecture or inducing oxidative stress (82, 83). Previous in vitro studies have shown that increased levels of ROS caused by IO have a severe impact on OBs proliferation, autophagy occurrence, differentiation, and mineralization in human, murine, and osteoblast-like cells (40, 84, 85). ROS may function by suppressing the PI3K/Akt/mTOR pathway, resulting in the stimulation of glycogen synthase kinase 3β (GSK-3β) (86, 87). Additionally, IO reduces canonical Wnt signaling, which further activates GSK-3β. The phosphorylation of GSK-3β is essential for the process of bone formation (osteogenesis). It directly controls the activity of RUNX2 or indirectly prevents the breakdown of β-catenin (88, 89). As a regulator of OBs differentiation and formation, Forkhead box transcription factor 1 (FOXO1) can directly interact with the promoter of RUNX2 to regulate its expression (90). FOX1 promotes bone formation by decreasing oxidative stress in OBs. ROS generated by IO may activate PI3K/Akt, leading to the inhibition of FOXO1 expression. This, in turn, impairs the survival of OBs (91, 92).

In addition, high levels of erythropoietin can directly affect the differentiation and mineralization of OBs progenitors, resulting in decreased bone density (93). This could be attributed to the overproduction of fibroblast growth factor (FGF)-5 in bone and BM erythroid cells, stimulated by erythropoietin through ERK23/1 and STAT2 pathways (94). Consequently, this results in the upregulation of FGF23 expression in bone and BM erythroid cells, leading to increased levels in BT patients and mice. Ultimately, this results in alterations in bone mineralization and disposition (94, 95). Inhibiting FGF23 signaling through carboxyl terminated FGF23 peptide is a safe and efficacious therapeutic approach to rescue bone mineralization and deposition in mice with β-thalassemia, normalizing the expression of niche factors and restoring HSCs function (94, 96). Additionally, OBs exposed to hyperglycemia show impaired function, such as decreased expression of ALP, GLA proteins, and OPN (9799).

Bone resorption, a common complication of thalassemia, has been shown to increase bone mineral density (BMD) in the femoral neck, lumbar spine, and forearm after two years of bisphosphonate treatment. After 12 months, the addition of zinc to one’s diet may lead to an increase in BMD specifically in the lumbar spine and hips (100). Moreover, supplementing zinc may improve iron-induced pancreatic exocrine and endocrine dysfunction (101). Current modalities for the management of osteoporosis in adults with TDT include inhibitors of bone remodeling such as bisphosphonates and denosumab as well as stimulators of bone formation, like teriparatide (28).

4 Bone microenvironment in case of bone marrow transplantation (BMT) patient

Poor transplantation function is a life-threatening complication that occurs after transplantation and has poor prognosis, limiting the success of BMT. The damaged bone marrow microenvironment is one of the important causes (102). In addition to the damage to the microenvironment itself, the treatment scheme used for transplantation can also damage and exacerbate the damage to the microenvironment. Cyclophosphamide(Cy) chemotherapy altered survival or proliferation of growth plate chondrocytes and metaphyseal osteoblastic cells and reduced heights of metaphyseal spongiosa trabecular bone, which may contribute to chemotherapy side effects of this drug on bone lengthening and bone mass accumulation (103). Cy reduces the number and differentiation of bone mesenchymal stem cells, as well as the formation and activity of osteoblasts. In addition, Cy inhibits the formation of osteoclasts by reducing their maturation and activity (104). Bone resorption is a common complication in thalassemia, and bisphosphonates may increase bone mineral density (BMD) in the femoral neck, lumbar spine, and forearm after two years of treatment. Zinc supplementation may increase BMD in the lumbar spine and hips after 12 months (100). In addition, supplementing zinc may improve iron induced pancreatic exocrine and endocrine dysfunction (101). Current modalities for the management of osteoporosis in adults with TDT include inhibitors of bone remodeling such as bisphosphonates and denosumab as well as stimulators of bone formation (e.g., teriparatide) (28). In addition, the myeloablative regimen can cause damage to BM EC, and the hematopoietic defects caused by damaged BM EC are positively correlated with ROS levels (105).

According to reports, atorvastatin is a widely used lipid-lowering drug in clinical practice, which improves the functional impairment of BM EPC in the body by downregulating the p38 MAPK pathway. In addition, NAC can reduce ROS levels in vitro and in vivo, while repairing damaged BM ECs to effectively promote hematopoietic reconstruction (105, 106). Due to the damage caused by Busulfan(BU) and Cy, researchers are constantly developing new protocols for pre-transplantation injection of BU or trisulfan (TREO) combined with thiacloprid (TT) and fludarabine (FLU) in patients with thalassemia, which have better overall survival and event free survival while reducing transplant failure (107). In addition, the combination of CY, intravenous BU, FLU, TT, and ATG (named NF-08-TM) has achieved excellent results in Mediterranean transplantation (108, 109). The RIC regimen combined with hydroxyurea (HU), alemtuzumab, FLU, melfaram (MEL), and TT is also superior to the BU-CY regimen (110). You can also add BIRC34, BIRC2, and NF- κ B expression to increase the cloning ability of CD34 cells.

5 Overall bone marrow pathological condition and scope of drug target based on BM condition

ROS is involved in the impaired function of MSCs, HSCs, OBs formation, and a series of complications in the BM microenvironment, which may be the second major factor affecting the BM microenvironment. The excess ROS can be neutralized by an efficient antioxidant system, which includes antioxidant enzymes and non-enzymatic molecules (111). Common antioxidant enzymes include superoxide dismutase (SOD), catalase, peroxidase (PRDXs), peroxiredoxin (Prxs), and glutathione peroxidase (GPXs) (111). However, the mechanism is inadequate in regulating ROS to reach normal levels in the injured BM microenvironment. At present, it can be regulated through other non-enzymatic antioxidant molecules such as glutathione, flavonoids, thioredoxin, and vitamins A, C, and E. These substances can be readily acquired as they are frequently found in plants (112). Resveratrol, found in the skin of fruits including grapes, blueberries, and raspberries, is a potent scavenger of ROS. It exerts a defensive influence on lipid peroxidation occurring in the plasma membrane and guards against DNA damage induced by ROS (113). Quercetin is a polyphenolic flavonoid found in apples, radishes, coriander, and cranberries, which can scavenge ROS and can be expressed through MAPK/ERK1/2, JAK/STAT and TRAIL, AMPK α 1/ASK1/p38, RAGE/PI3K/AKT/mTOR axis, HMGB1, and NF- κ B. Nrf2 and other signaling pathways regulate cell state (114). N-acetylcysteine (NAC) is an antioxidant derived from a small amino acid with a low molecular weight, which allows it to be rapidly delivered to the cytoplasm (115). Moreover, NAC has been shown to greatly enhance bone healing and its ability to promote bone formation is evident. However, further research is required to fully understand the particular mechanism behind this benefit (115). Other sources of dietary antioxidants include rutin, anthocyanins, chlorogenic acid, quinic acid, caffeic acid, etc (116). These substances all regulate the activity of endogenous oxidase systems and their related proteins, preventing oxidative damage to organelles, proteins, nucleic acids, and lipids (117). Although there is evidence to support these effects at this time, to avoid adverse reactions, it is important to consider the dose when using these substances. Patients with thalassemia should be advised to add foods high in these compounds to their diet in addition to receiving therapy.

6 Thalassemia and hematological tumors

In recent years, with the continuous improvement of treatment plans, the survival rate of patients with thalassemia has been significantly improved. However, more complications have emerged, such as the rising incidence of liver cancer (118). A comprehensive longitudinal research conducted in Taiwan has revealed that individuals with thalassemia have a significantly elevated total incidence rate of cancer, reaching 52%. Furthermore, the chance of developing lymphoma or leukemia was found to be 5.32 times greater in these patients. People with transfusion-dependent thalassemia have a 9.31-fold higher risk of developing hematological malignancies compared to people who do not require transfusions (119). Reported hematological tumors include hematological diseases such as acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic myeloid leukemia (CML), essential thrombocythaemia (ET), Hodgkin lymphoma (HL), multiple myeloma (MM), myeloproliferative neoplasm (MPN), non-Hodgkin lymphoma (NHL) and Diffuse large B cell lymphoma (DLBCL), etc (120123). The increased tumor risk in patients with thalassemia may be related to oxidative damage secondary to iron accumulation, immune abnormalities, viral infections, IO, hydroxyurea use, and BM stimulation caused by chronic anemia (124, 125). Furthermore, thalassemia can manifest after the development of a tumor. The loss of α-globin gene clusters and the inactivation of somatic mutations in the trans-acting chromatin-associated protein ATRX usually lead to a significant decrease in α-globin gene expression (126). This type of acquired thalassemia is also seen in Hodgkin’s lymphoma, Juvenile Myelomonocytic Leukemia, and others (127, 128). However, the mechanism related to thalassemia and hematologic tumorigenesis is still unclear, which needs more research to reveal.

7 Other

Endothelial cell activation and dysfunction are confirmed in BT, mostly due to the inhibitory effect of ADMA on NO (nitric oxide) production and the accumulation of iron, which disrupts endothelial function (129131). Endothelial cell apoptosis in the circulation involves the mitogen-activated protein kinases/Jun N-terminal Kinase (MAPK/JNK) signaling pathway (132). Currently, ongoing studies on BT endothelium are mostly centered around investigating the relationship between direct or indirect measures of IO (namely serum ferritin, transfusion burden, and MRI results) and outcome parameters (133). Furthermore, several research investigations have demonstrated an elevation in levels of adhesion molecules (intracellular adhesion molecule-1, slCAM-1, sVCAM-1, E-selectin, and P-selectin) and inflammatory factors (IL-6 and IL-1β) in individuals with thalassemia, along with an increase in tissue factor levels (134, 135). Long-term exposure to compromised erythropoiesis in the BM microenvironment was not directly considered. According to studies, atorvastatin, a widely used lipid-lowering drug in clinical practice, improves the functional impairment of BM EPC in the body by downregulating the p38 MAPK pathway. Moreover, NAC was also found to reduce ROS levels in vitro and in vivo, while repairing damaged BM ECs to effectively promote hematopoietic reconstruction (105, 106).

Macrophages within erythrocyte islands are central to the normal differentiation and development of erythrocytes (136). After being cultured with murine macrophages, it was discovered that the erythrocyte precursors extracted from BT BM were phagocytosed by the macrophages (137). Even while erythrocyte precursors derived from healthy persons were likewise phagocytosed, there was a significant rise in the activity of BT BM macrophages (137, 138). This may suggest that macrophage phagocytosis is enhanced in BT BM. Marrow adipose tissue(MAT), one of the main components of the BM stroma, plays a crucial role in maintaining hematopoietic function (139). However, the MAT was decreased in the BM of BT individuals with inefficient erythropoiesis, particularly in the BM fat fraction (BMFF) in red and yellow BM regions. This indicates that the transformation process of the BM was hindered (35, 140).

8 Conclusion

In summary, the review focused on the compromised BM microenvironment in patients with BT and the subsequent chain of events, which has an adverse impact on the patient. Hence, it is imperative to focus on the compromised BM microenvironment of patients, particularly IO, ROS, and bone resorption. Patients should get iron chelation therapy, antioxidant therapy, hypoglycemic therapy, and therapy to improve bone mineral density at an appropriate time.

Author contributions

ZL: Funding acquisition, Methodology, Project administration, Writing – original draft. XY: Supervision, Writing – review & editing. JZ: Formal analysis, Supervision, Writing – original draft. JY: Conceptualization, Formal analysis, Writing – review & editing. JN: Conceptualization, Visualization, Writing – review & editing. YW: Funding acquisition, Project administration, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by grants from the National Natural Science Foundation of China (82070173 and 81460678). Yunnan Applied Basic Research Projects (Joint fund for applied basic research of Kunming Medical University, Yunnan Provincial Department of science and technology)(2018FE001(-156), 2017FE468(-105)). Candidate Talents Training Fund of Yunnan Province (H2017012). Yunnan Province Basic Research Excellent Youth Project (202101AW070017). Doctoral Research Fund of Yunnan First People’s Hospital (KHBS-2020–007). Open Project of Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment (2021LCZXXF-XY09 and 2022LCZXKF-XY01). Yunnan Provincial Department of Science and Technology—Kunming Medical University Joint Special Project on Applied Basic Research (202301AY070001–064 and 202201AY070001–232). Top Experts training Project for the Academy and Technology in Yunnan province (Grant No. 202105AC160030). Famous doctor project of Xingdian talent plan in Yunnan province (XDYC- MY-2022-0005).

Acknowledgments

We are grateful to our lab members and collaborators for their assistance with the experiments and in the preparation of the manuscript.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Rivella S. beta-thalassemias: paradigmatic diseases for scientific discoveries and development of innovative therapies. Haematologica. (2015) 100:418–30. doi: 10.3324/haematol.2014.114827

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Rivella S. Iron metabolism under conditions of ineffective erythropoiesis in β-thalassemia. Blood. (2019) 133:51–8. doi: 10.1182/blood-2018-07-815928

PubMed Abstract | CrossRef Full Text | Google Scholar

3. McGann PT, Nero AC, Ware RE. Clinical features of beta-thalassemia and sickle cell disease. Adv Exp Med Biol. (2017) 1013:1–26. doi: 10.1007/978-1-4939-7299-9_1

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Carsote M, Vasiliu C, Trandafir AI, Albu SE, Dumitrascu MC, Popa A, et al. New entity-thalassemic endocrine disease: major beta-thalassemia and endocrine involvement. Diagnostics (Basel). (2022) 12. doi: 10.3390/diagnostics12081921

CrossRef Full Text | Google Scholar

5. Yuan J, Kannan R, Shinar E, Rachmilewitz E, Low P. Isolation, characterization, and immunoprecipitation studies of immune complexes from membranes of beta-thalassemic erythrocytes. Blood. (1992) 79:3007–13. doi: 10.1182/blood.V79.11.3007.3007

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Matte A, De Falco L, Iolascon A, Mohandas N, An X, Siciliano A, et al. The interplay between peroxiredoxin-2 and nuclear factor-Erythroid 2 is important in limiting oxidative mediated dysfunction in beta-Thalassemic erythropoiesis. Antioxid Redox Signal. (2015) 23:1284–97. doi: 10.1089/ars.2014.6237

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Arlet JB, Ribeil JA, Guillem F, Negre O, Hazoume A, Marcion G, et al. HSP70 sequestration by free alpha-globin promotes ineffective erythropoiesis in beta-thalassaemia. Nature. (2014) 514:242–6. doi: 10.1038/nature13614

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Dussiot M, Maciel TT, Fricot A, Chartier C, Negre O, Veiga J, et al. An activin receptor IIA ligand trap corrects ineffective erythropoiesis in beta-thalassemia. Nat Med. (2014) 20:398–407. doi: 10.1038/nm.3468

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Suragani RN, Cadena SM, Cawley SM, Sako D, Mitchell D, Li R, et al. Transforming growth factor-beta superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat Med. (2014) 20:408–14. doi: 10.1038/nm.3512

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Gluba-Brzozka A, Franczyk B, Rysz-Gorzynska M, Rokicki R, Koziarska-Rosciszewska M, Rysz J. Pathomechanisms of immunological disturbances in beta-thalassemia. Int J Mol Sci. (2021) 22. doi: 10.3390/ijms22189677

CrossRef Full Text | Google Scholar

11. Xu B, Hu R, Liang Z, Chen T, Chen J, Hu Y, et al. Metabolic regulation of the bone marrow microenvironment in leukemia. Blood Rev. (2021) 48:100786. doi: 10.1016/j.blre.2020.100786

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Wei Q, Frenette PS. Niches for hematopoietic stem cells and their progeny. Immunity. (2018) 48:632–48. doi: 10.1016/j.immuni.2018.03.024

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kumar R, Godavarthy PS, Krause DS. The bone marrow microenvironment in health and disease at a glance. J Cell Sci. (2018) 131. doi: 10.1242/jcs.201707

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Wu C-H, Weng T-F, Li J-P, Wu K-H. Biology and therapeutic properties of mesenchymal stem cells in leukemia. Int J Mol Sci. (2024) 25. doi: 10.3390/ijms25052527

CrossRef Full Text | Google Scholar

15. Quagliano A, Gopalakrishnapillai A, Barwe SP. Tetraspanins set the stage for bone marrow microenvironment–induced chemoprotection in hematologic Malignancies. Blood Adv. (2023) 7:4403–13. doi: 10.1182/bloodadvances.2023010476

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Nyamondo K, Wheadon H. Micro-environment alterations through time leading to myeloid Malignancies. BRIT J Pharmacol. (2024) 181:283–94. doi: 10.1111/bph.15924

CrossRef Full Text | Google Scholar

17. Aprile A, Gulino A, Storto M, Villa I, Beretta S, Merelli I, et al. Hematopoietic stem cell function in β-thalassemia is impaired and is rescued by targeting the bone marrow niche. Blood. (2020) 136:610–22. doi: 10.1182/blood.2019002721

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Look R, Koh H, Hamburg S, Lim S, Waxman A, Jochelson M, et al. Technetium-99m sestamibi scanning reflects marrow activity in a β-Thalassemia patient treated with an allogeneic bone marrow transplant. Hematology. (1996) 1:173–6. doi: 10.1080/10245332.1996.11746302

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Aksoy C, Guliyev A, Kilic E, Uckan D, Severcan F. Bone marrow mesenchymal stem cells in patients with beta thalassemia major: molecular analysis with attenuated total reflection-Fourier transform infrared spectroscopy study as a novel method. Stem Cells Dev. (2012) 21:2000–11. doi: 10.1089/scd.2011.0444

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Bathija A, Davis S, Trubowitz S. Marrow adipose tissue: response to erythropoiesis. Am J Hematol. (1978) 5:315–21. doi: 10.1002/ajh.2830050406

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Ismail UN, Azlan CA, Khairullah S, Azman RR, Omar NF, Md Shah MN, et al. Marrow fat content and composition in β-thalassemia: A study using 1H-MRS. J Magnetic Resonance Imaging. (2020) 53:190–8. doi: 10.1002/jmri.27294

CrossRef Full Text | Google Scholar

22. Ponnikorn S, Mongkolrob R, Klongthalay S, Roytrakul S, Srisanga K, Tungpradabkul S, et al. Comparative proteome-wide analysis of bone marrow microenvironment of β-thalassemia/hemoglobin E. Proteomes. (2019) 7. doi: 10.3390/proteomes7010008

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Khan M, Iftikhar F, Khan T, Danish A, Shamsi T, Musharraf S, et al. IVS I-5 (G > C) is associated with changes to the RBC membrane lipidome in response to hydroxyurea treatment in β-thalassemia patients. Mol Omics. (2022) 18:534–44. doi: 10.1039/D2MO00008C

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Khalid A, Siddiqui A, Ansari S, Musharraf S. Reflection of treatment proficiency of hydroxyurea treated β-thalassemia serum samples through nuclear magnetic resonance based metabonomics. Sci Rep. (2019) 9:2041. doi: 10.1038/s41598-019-38823-0

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Iqbal A, Siddiqui A, Huang J, Ansari S, Musharraf S. Impact of hydroxyurea therapy on serum fatty acids of β-thalassemia patients. Metabolomics. (2018) 14:27. doi: 10.1007/s11306-018-1325-0

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Taneera J, Mahgoub E, Qannita R, Alalami A, Shehadat O, Youssef M, et al. β-thalassemia and diabetes mellitus: current state and future directions. Horm Metab Res. (2024) 56:272–8. doi: 10.1055/a-2185-5073

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Santarone S, Angelini S, Natale A, Vaddinelli D, Spadano R, Casciani P, et al. Survival and late effects of hematopoietic cell transplantation in patients with thalassemia major. Bone Marrow Transplant. (2022) 57:1689–97. doi: 10.1038/s41409-022-01786-4

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Tzoulis P, Yavropoulou M, Banchev A, Modeva I, Daar S, De Sanctis V. Recent advancements in glucose dysregulation and pharmacological management of osteoporosis in transfusion-dependent thalassemia (TDT): an update of ICET-A (International Network of Clinicians for Endocrinopathies in Thalassemia and Adolescence Medicine). Acta BioMed. (2023) 94:e2023178. doi: 10.23750/abm.v94i3.14805

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Meshkani S, Mahdian D, Abbaszadeh-Goudarzi K, Abroudi M, Dadashizadeh G, Lalau J, et al. Metformin as a protective agent against natural or chemical toxicities: a comprehensive review on drug repositioning. J Endocrinol Invest. (2020) 43:1–19. doi: 10.1007/s40618-019-01060-3

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Halabi A, Sen J, Huynh Q, Marwick TH. Metformin treatment in heart failure with preserved ejection fraction: a systematic review and meta-regression analysis. Cardiovasc Diabetol. (2020) 19:124. doi: 10.1186/s12933-020-01100-w

PubMed Abstract | CrossRef Full Text | Google Scholar

31. De Broe M, Kajbaf F, Lalau J. Renoprotective effects of metformin. Nephron. (2018) 138:261–74. doi: 10.1159/000481951

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Semo D, Obergassel J, Dorenkamp M, Hemling P, Strutz J, Hiden U, et al. The Sodium-Glucose Co-Transporter 2 (SGLT2) Inhibitor Empagliflozin Reverses Hyperglycemia-Induced Monocyte and Endothelial Dysfunction Primarily through Glucose Transport-Independent but Redox-Dependent Mechanisms. J Clin Med. (2023) 12. doi: 10.3390/jcm12041356

CrossRef Full Text | Google Scholar

33. Bode D, Semmler L, Wakula P, Hegemann N, Primessnig U, Beindorff N, et al. Dual SGLT-1 and SGLT-2 inhibition improves left atrial dysfunction in HFpEF. Cardiovasc Diabetol. (2021) 20:7. doi: 10.1186/s12933-020-01208-z

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Gliozzi M, Macrì R, Coppoletta AR, Musolino V, Carresi C, Scicchitano M, et al. From diabetes care to heart failure management: A potential therapeutic approach combining SGLT2 inhibitors and plant extracts. Nutrients. (2022) 14. doi: 10.3390/nu14183737

CrossRef Full Text | Google Scholar

35. Lithanatudom P, Leecharoenkiat A, Wannatung T, Svasti S, Fucharoen S, Smith DR. A mechanism of ineffective erythropoiesis in -thalassemia/Hb E disease. Haematologica. (2009) 95:716–23. doi: 10.3324/haematol.2009.015701

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Taher A, Saliba AN, Harb A. Iron chelation therapy in transfusion-dependent thalassemia patients: current strategies and future directions. J Blood Med. (2015) 6. doi: 10.2147/JBM.S72463

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Bou-Fakhredin R, Bazarbachi A-H, Chaya B, Sleiman J, Cappellini M, Taher A. Iron overload and chelation therapy in non-transfusion dependent thalassemia. Int J Mol Sci. (2017) 18. doi: 10.3390/ijms18122778

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Taher A, Saliba A. Iron overload in thalassemia: different organs at different rates. Hematol-Am Soc Hemat. (2017) 2017:265–71. doi: 10.1182/asheducation-2017.1.265

CrossRef Full Text | Google Scholar

39. Crippa S, Rossella V, Aprile A, Silvestri L, Rivis S, Scaramuzza S, et al. Bone marrow stromal cells from β-thalassemia patients have impaired hematopoietic supportive capacity. J Clin Invest. (2019) 129:1566–80. doi: 10.1172/JCI123191

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Piriyakhuntorn P, Tantiworawit A, Phimphilai M, Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. Impact of iron overload on bone remodeling in thalassemia. Arch Osteoporosis. (2020) 15:143. doi: 10.1007/s11657-020-00819-z

CrossRef Full Text | Google Scholar

41. Song X, Xie Y, Kang R, Hou W, Sun X, Epperly MW, et al. FANCD2 protects against bone marrow injury from ferroptosis. Biochem Biophys Res Commun. (2016) 480:443–9. doi: 10.1016/j.bbrc.2016.10.068

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Gammella E, Recalcati S, Cairo G. Dual role of ROS as signal and stress agents: iron tips the balance in favor of toxic effects. Oxid Med Cell Longevity. (2016) 2016:1–9. doi: 10.1155/2016/8629024

CrossRef Full Text | Google Scholar

43. Bou-Fakhredin R, Dia B, Ghadieh H, Rivella S, Cappellini M, Eid A, et al. CYP450 mediates reactive oxygen species production in a mouse model of β-thalassemia through an increase in 20-HETE activity. Int J Mol Sci. (2021) 22. doi: 10.3390/ijms22031106

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Saensuwanna A, Penglong T, Srinoun K. Upregulation of miR-214 Mediates Oxidative Stress in Hb H Disease via Targeting of ATF4. HEMOGLOBIN. (2021) 45:197–202. doi: 10.1080/03630269.2021.1941081

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Cairo G, Recalcati S. Iron-regulatory proteins: molecular biology and pathophysiological implications. Expert Rev Mol Med. (2007) 9:1–13. doi: 10.1017/S1462399407000531

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Tsamesidis I, Fozza C, Vagdatli E, Kalpaka A, Cirotto C, Pau M, et al. Total antioxidant capacity in Mediterranean β-thalassemic patients. Adv Clin Exp Med. (2017) 26:789–93. doi: 10.17219/acem/63746

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Mezey E, Zhang Y, Zhai W, Zhao M, Li D, Chai X, et al. Effects of iron overload on the bone marrow microenvironment in mice. PloS One. (2015) 10:e0120219. doi: 10.1371/journal.pone.0120219

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Urao N, Ushio-Fukai M. Redox regulation of stem/progenitor cells and bone marrow niche. Free Radical Biol Med. (2013) 54:26–39. doi: 10.1016/j.freeradbiomed.2012.10.532

CrossRef Full Text | Google Scholar

49. Panwar N, Gomber S, Dewan P, Kumar R. Pulmonary dysfunction in transfusion-dependent thalassemia and response to intensive chelation therapy. Indian Pediatr. (2022) 59:451–4. doi: 10.1007/s13312-022-2533-2

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Casu C, Pettinato M, Liu A, Aghajan M, Lo Presti V, Lidonnici M, et al. Correcting β-thalassemia by combined therapies that restrict iron and modulate erythropoietin activity. Blood. (2020) 136:1968–79. doi: 10.1182/blood.2019004719

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Paul A, Thomson V, Refat M, Al-Rawahi B, Taher A, Nadar S. Cardiac involvement in beta-thalassaemia: current treatment strategies. Postgrad Med. (2019) 131:261–7. doi: 10.1080/00325481.2019.1608071

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Ahmed S, Soliman A, De Sanctis V, Alaaraj N, Alyafei F, Hamed N, et al. A short review on growth and endocrine long-term complications in children and adolescents with β-thalassemia major: conventional treatment versus hematopoietic stem cell transplantation. Acta BioMed. (2022) 93:e2022290. doi: 10.23750/abm.v93i4.13331

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Inati A, Kahale M, Sbeiti N, Cappellini M, Taher A, Koussa S, et al. One-year results from a prospective randomized trial comparing phlebotomy with deferasirox for the treatment of iron overload in pediatric patients with thalassemia major following curative stem cell transplantation. Pediatr Blood Cancer. (2017) 64:188–96. doi: 10.1002/pbc.26213

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Kupesiz F, Sivrice C, Akinel A, Kintrup G, Guler E, Kupesiz A. Efficacy and safety of iron chelation therapy after allogeneic hematopoietic stem cell transplantation in pediatric thalassemia patients: A retrospective observational study. J Pediat Hematol Onc. (2022) 44:e26–34. doi: 10.1097/MPH.0000000000002328

CrossRef Full Text | Google Scholar

55. Yesilipek MA, Karasu G, Kaya Z, Kuskonmaz BB, Uygun V, Dag I, et al. A Phase II, Multicenter, Single-Arm Study to Evaluate the Safety and Efficacy of Deferasirox after Hematopoietic Stem Cell Transplantation in Children with beta-Thalassemia Major. Biol Blood Marrow Transplant. (2018) 24:613–8. doi: 10.1016/j.bbmt.2017.11.006

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Cappellini M, Motta I. New therapeutic targets in transfusion-dependent and -independent thalassemia. Hematol-Am Soc Hemat. (2017) 2017:278–83. doi: 10.1182/asheducation-2017.1.278

CrossRef Full Text | Google Scholar

57. Casu C, Nemeth E, Rivella S. Hepcidin agonists as therapeutic tools. BLOOD. (2018) 131:1790–4. doi: 10.1182/blood-2017-11-737411

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Hunter Z, Treon S. Epigenomics in waldenström macroglobulinemia. BLOOD. (2020) 136:527–9. doi: 10.1182/blood.2020006244

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Hua P, Roy N, de la Fuente J, Wang G, Thongjuea S, Clark K, et al. Single-cell analysis of bone marrow-derived CD34+ cells from children with sickle cell disease and thalassemia. BLOOD. (2019) 134:2111–5. doi: 10.1182/blood.2019002301

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Nilsson S, Johnston H, Whitty G, Williams B, Webb R, Denhardt D, et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood. (2005) 106:1232–9. doi: 10.1182/blood-2004-11-4422

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Crippa S, Conti A, Vavassori V, Ferrari S, Beretta S, Rivis S, et al. Mesenchymal stromal cells improve the transplantation outcome of CRISPR-Cas9 gene-edited human HSPCs. Mol Ther. (2023) 31:230–48. doi: 10.1016/j.ymthe.2022.08.011

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Rostami T, Kasaeian A, Maleki N, Nikbakht M, Kiumarsi A, Tavangar SM, et al. The effect of bone marrow-derived mesenchymal stem cell co-transplantation with hematopoietic stem cells on liver fibrosis alleviation and survival in patients with class III β-thalassemia major. Stem Cell Res Ther. (2021) 12:213. doi: 10.1186/s13287-021-02242-8

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Rostami T, Maleki N, Kasaeian A, Nikbakht M, Kiumarsi A, Asadollah Mousavi S, et al. Co-transplantation of bone marrow-derived mesenchymal stem cells with hematopoietic stem cells does not improve transplantation outcome in class III beta-thalassemia major: A prospective cohort study with long-term follow-up. Pediatr Transplant. (2021) 25:e13905. doi: 10.1111/petr.13905

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Preciado S, Muntión S, Sánchez-Guijo F. Improving hematopoietic engraftment: Potential role of mesenchymal stromal cell-derived extracellular vesicles. Stem Cells. (2021) 39:26–32. doi: 10.1002/stem.3278

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Preciado S, Muntión S, Corchete LA, Ramos TL, Torre AG, Osugui L, et al. The incorporation of extracellular vesicles from mesenchymal stromal cells into CD34+ Cells increases their clonogenic capacity and bone marrow lodging ability. Stem Cells. (2019) 37:1357–68. doi: 10.1002/stem.3032

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Wen S, Dooner M, Cheng Y, Papa E, Del Tatto M, Pereira M, et al. Mesenchymal stromal cell-derived extracellular vesicles rescue radiation damage to murine marrow hematopoietic cells. Leukemia. (2016) 30:2221–31. doi: 10.1038/leu.2016.107

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. (2006) 25:977–88. doi: 10.1016/j.immuni.2006.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Tamma R, Ribatti D. Bone niches, hematopoietic stem cells, and vessel formation. Int J Mol Sci. (2017) 18. doi: 10.3390/ijms18010151

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Pricola KL, Kuhn NZ, Haleem-Smith H, Song Y, Tuan RS. Interleukin-6 maintains bone marrow-derived mesenchymal stem cell stemness by an ERK1/2-dependent mechanism. J Cell Biochem. (2009) 108:577–88. doi: 10.1002/jcb.22289

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Genovese P, Schiroli G, Escobar G, Di Tomaso T, Firrito C, Calabria A, et al. Targeted genome editing in human repopulating haematopoietic stem cells. Nature. (2014) 510:235–40. doi: 10.1038/nature13420

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Hongeng S, Wongborisuth C, Disthabanchong S, Pakakasama S, Mahachoklertwattana P, Ongphiphadhanakul B, et al. Use of in vivo gene expression of isolated bone marrow mesenchymal stromal cells to study the pathophysiology of osteoporosis in patients with severe thalassemia. J Pediat Hematol Onc. (2011) 33:179–84. doi: 10.1097/MPH.0b013e318205e24d

CrossRef Full Text | Google Scholar

72. Almalki S, Agrawal D. Key transcription factors in the differentiation of mesenchymal stem cells. Differentiation. (2016) 92:41–51. doi: 10.1016/j.diff.2016.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Gomathi K, Akshaya N, Srinaath N, Moorthi A, Selvamurugan N. Regulation of Runx2 by post-translational modifications in osteoblast differentiation. Life Sci. (2020) 245:117389. doi: 10.1016/j.lfs.2020.117389

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Vogiatzi MG, Tsay J, Verdelis K, Rivella S, Grady RW, Doty S, et al. Changes in bone microarchitecture and biomechanical properties in the th3 thalassemia mouse are associated with decreased bone turnover and occur during the period of bone accrual. Calcified Tissue Int. (2010) 86:484–94. doi: 10.1007/s00223-010-9365-0

CrossRef Full Text | Google Scholar

75. Bowers M, Zhang B, Ho Y, Agarwal P, Chen C, Bhatia R. Osteoblast ablation reduces normal long-term hematopoietic stem cell self-renewal but accelerates leukemia development. Blood. (2015) 125:2678–88. doi: 10.1182/blood-2014-06-582924

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Bonnet D, Ariza-McNaughton L, Foster K, Abarrategi A, Passaro D. Bioengineering of humanized bone marrow microenvironments in mouse and their visualization by live imaging. J Visualized Experiments. (2017) 126). doi: 10.3791/55914

CrossRef Full Text | Google Scholar

77. Bhardwaj A, Swe K, Sinha N, Osunkwo I. Treatment for osteoporosis in people with ß-thalassaemia. Cochrane Database Syst Rev. (2016) 3:CD010429. doi: 10.1002/14651858.CD010429.pub2

PubMed Abstract | CrossRef Full Text | Google Scholar

78. De Sanctis V, Soliman AT, Elsefdy H, Soliman N, Bedair E, Fiscina B, et al. Bone disease in β thalassemia patients: past, present and future perspectives. Metabolism. (2018) 80:66–79. doi: 10.1016/j.metabol.2017.09.012

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Piga A. Impact of bone disease and pain in thalassemia. HEMATOL-AM Soc HEMAT. (2017) 2017:272–7. doi: 10.1182/asheducation-2017.1.272

CrossRef Full Text | Google Scholar

80. Stier S, Ko Y, Forkert R, Lutz C, Neuhaus T, Grünewald E, et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J Exp Med. (2005) 201:1781–91. doi: 10.1084/jem.20041992

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Kasai K, Hori M, Goodman W. Characterization of the transferrin receptor in UMR-106-01 osteoblast-like cells. Endocrinolgy. (1990) 126:1742–9. doi: 10.1210/endo-126-3-1742

CrossRef Full Text | Google Scholar

82. Isidori A, Borin L, Elli E, Latagliata R, Martino B, Palumbo G, et al. Iron toxicity - Its effect on the bone marrow. Blood Rev. (2018) 32:473–9. doi: 10.1016/j.blre.2018.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Lertsuwan K, Nammultriputtar K, Nanthawuttiphan S, Phoaubon S, Lertsuwan J, Thongbunchoo J, et al. Ferrous and ferric differentially deteriorate proliferation and differentiation of osteoblast-like UMR-106 cells. BIOMETALS. (2018) 31:873–89. doi: 10.1007/s10534-018-0130-6

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Balogh E, Tolnai E, Nagy B, Nagy B, Balla G, Balla J, et al. Iron overload inhibits osteogenic commitment and differentiation of mesenchymal stem cells via the induction of ferritin. Biochim Biophys Acta. (2016) 1862:1640–9. doi: 10.1016/j.bbadis.2016.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Xu G, Li X, Zhu Z, Wang H, Bai X. Iron overload induces apoptosis and cytoprotective autophagy regulated by ROS generation in mc3t3-E1 cells. Biol Trace ELEM Res. (2021) 199:3781–92. doi: 10.1007/s12011-020-02508-x

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Ke J, Cen W, Zhou X, Li Y, Kong W, Jiang J. Iron overload induces apoptosis of murine preosteoblast cells via ROS and inhibition of AKT pathway. Oral Dis. (2017) 23:784–94. doi: 10.1111/odi.12662

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Zhang P, Wang S, Wang L, Shan B, Zhang H, Yang F, et al. Hepcidin is an endogenous protective factor for osteoporosis by reducing iron levels. J Mol Endocrinol. (2018) 60:297–306. doi: 10.1530/JME-17-0301

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Heisenberg C-P, Kugimiya F, Kawaguchi H, Ohba S, Kawamura N, Hirata M, et al. GSK-3β Controls osteogenesis through regulating runx2 activity. PloS One. (2007) 2:e837. doi: 10.1371/journal.pone.0000837

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Tsay J, Yang Z, Ross F, Cunningham-Rundles S, Lin H, Coleman R, et al. Bone loss caused by iron overload in a murine model: importance of oxidative stress. Blood. (2010) 14(2582-9):2582–9. doi: 10.1182/blood-2009-12-260083

CrossRef Full Text | Google Scholar

90. Teixeira CC, Liu Y, Thant LM, Pang J, Palmer G, Alikhani M. Foxo1, a novel regulator of osteoblast differentiation and skeletogenesis. J Biol Chem. (2010) 285:31055–65. doi: 10.1074/jbc.M109.079962

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Xia D, Wu J, Xing M, Wang Y, Zhang H, Xia Y, et al. Iron overload threatens the growth of osteoblast cells via inhibiting the PI3K/AKT/FOXO3a/DUSP14 signaling pathway. J Cell Physiol. (2019) 234:15668–77. doi: 10.1002/jcp.28217

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Rached M-T, Kode A, Xu L, Yoshikawa Y, Paik J-H, DePinho RA, et al. FoxO1 is a positive regulator of bone formation by favoring protein synthesis and resistance to oxidative stress in osteoblasts. Cell Metab. (2010) 11:147–60. doi: 10.1016/j.cmet.2010.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Rauner M, Franke K, Murray M, Singh R, Hiram-Bab S, Platzbecker U, et al. Increased EPO levels are associated with bone loss in mice lacking PHD2 in EPO-Producing cells. J Bone MINER Res. (2016) 31:1877–87. doi: 10.1002/jbmr.2857

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Aprile A, Raggi L, Bolamperti S, Villa I, Storto M, Morello G, et al. Inhibition of FGF23 is a therapeutic strategy to target hematopoietic stem cell niche defects in β-thalassemia. Sci Transl Med. (2023) 15:eabq3679. doi: 10.1126/scitranslmed.abq3679

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Stefanopoulos D, Nasiri-Ansari N, Dontas I, Vryonidou A, Galanos A, Psaridi L, et al. Fibroblast growth factor 23 (FGF23) and klotho protein in beta-thalassemia. Hormone Metab Res. (2020) 52:194–201. doi: 10.1055/a-1104-5326

CrossRef Full Text | Google Scholar

96. Saki F, Salehifar A, Kassaee SR, Omrani GR. Association of vitamin D and FGF23 with serum ferritin in hypoparathyroid thalassemia: a case control study. BMC Nephrol. (2020) 21:482. doi: 10.1186/s12882-020-02101-3

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Hamann C, Goettsch C, Mettelsiefen J, Henkenjohann V, Rauner M, Hempel U, et al. Delayed bone regeneration and low bone mass in a rat model of insulin-resistant type 2 diabetes mellitus is due to impaired osteoblast function. Am J Physiol-Endoc M. (2011) 301:E1220–1228. doi: 10.1152/ajpendo.00378.2011

CrossRef Full Text | Google Scholar

98. Fujii H, Hamada Y, Fukagawa M. Bone formation in spontaneously diabetic Torii-newly established model of non-obese type 2 diabetes rats. BONE. (2008) 42:372–9. doi: 10.1016/j.bone.2007.10.007

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Charoenphandhu N, Aeimlapa R, Sooksawanwit S, Thongbunchoo J, Teerapornpuntakit J, Svasti S, et al. Responses of primary osteoblasts and osteoclasts from hemizygous β-globin knockout thalassemic mice with elevated plasma glucose to 1,25-dihydroxyvitamin D3. Sci Rep. (2019) 9:13963. doi: 10.1038/s41598-019-50414-7

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Bhardwaj A, Swe KMM, Sinha NK. Treatment for osteoporosis in people with beta-thalassaemia. Cochrane Database Systematic Rev. (2023) 2023:CD010429. doi: 10.1002/14651858.CD010429.pub3

CrossRef Full Text | Google Scholar

101. Mousa SO, Abd Alsamia EM, Moness HM, Mohamed OG. The effect of zinc deficiency and iron overload on endocrine and exocrine pancreatic function in children with transfusion-dependent thalassemia: a cross-sectional study. BMC Pediatr. (2021) 21:468. doi: 10.1186/s12887-021-02940-5

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Leventogiannis K, Kyriazopoulou E, Antonakos N, Kotsaki A, Tsangaris I, Markopoulou D, et al. Toward personalized immunotherapy in sepsis: The PROVIDE randomized clinical trial. Cell Rep Med. (2022) 3:100817. doi: 10.2139/ssrn.4054844

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Xian CJ, Cool JC, van Gangelen J, Foster BK, Howarth GS. Effects of etoposide and cyclophosphamide acute chemotherapy on growth plate and metaphyseal bone in rats. Cancer Biol Ther. (2014) 6:170–7. doi: 10.4161/cbt.6.2.3576

CrossRef Full Text | Google Scholar

104. Zhao D, Wang C, Zhao Y, Shu B, Jia Y, Liu S, et al. Cyclophosphamide causes osteoporosis in C57BL/6 male mice: suppressive effects of cyclophosphamide on osteoblastogenesis and osteoclastogenesis. Oncotarget. (2017) 8:98163–83. doi: 10.18632/oncotarget.21000

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Kong Y, Wang Y, Zhang Y-Y, Shi M-M, Mo X-D, Sun Y-Q, et al. Prophylactic oral NAC reduced poor hematopoietic reconstitution by improving endothelial cells after haploidentical transplantation. Blood Adv. (2019) 3:1303–17. doi: 10.1182/bloodadvances.2018029454

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Shi M-M, Kong Y, Song Y, Sun Y-Q, Wang Y, Zhang X-H, et al. Atorvastatin enhances endothelial cell function in posttransplant poor graft function. Blood. (2016) 128:2988–99. doi: 10.1182/blood-2016-03-702803

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Bernardo ME, Piras E, Vacca A, Giorgiani G, Zecca M, Bertaina A, et al. Allogeneic hematopoietic stem cell transplantation in thalassemia major: results of a reduced-toxicity conditioning regimen based on the use of treosulfan. Blood. (2012) 120:473–6. doi: 10.1182/blood-2012-04-423822

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Eapen M, Walters MC, Smith AR, Shenoy S, Kasow K, Hanna R, et al. Related and unrelated donor transplantation for β-thalassemia major: results of an international survey. Blood Adv. (2019) 3:2562–70. doi: 10.1182/bloodadvances.2019000291

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Li C, Wu X, Feng X, He Y, Liu H, Pei F, et al. A novel conditioning regimen improves outcomes in β-thalassemia major patients using unrelated donor peripheral blood stem cell transplantation. Blood. (2012) 120:3875–81. doi: 10.1182/blood-2012-03-417998

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Shenoy S, Walters MC, Ngwube A, Soni S, Jacobsohn D, Chaudhury S, et al. Unrelated donor transplantation in children with thalassemia using reduced-Intensity conditioning: the URTH trial. Biol Blood Marrow Transplant. (2018) 24:1216–22. doi: 10.1016/j.bbmt.2018.01.023

PubMed Abstract | CrossRef Full Text | Google Scholar

111. D’Autréaux B, Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. (2007) 8:813–24. doi: 10.1038/nrm2256

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Huang J, Zhao Y, Zhao K, Yin K, Wang S. Function of reactive oxygen species in myeloid-derived suppressor cells. Front Immunol. (2023) 14. doi: 10.3389/fimmu.2023.1226443

CrossRef Full Text | Google Scholar

113. Truong V, Jun M, Jeong W. Role of resveratrol in regulation of cellular defense systems against oxidative stress. BIOFACTORS. (2018) 44:36–49. doi: 10.1002/biof.1399

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Biswas P, Dey D, Biswas PK, Rahaman TI, Saha S, Parvez A, et al. A comprehensive analysis and anti-cancer activities of quercetin in ROS-mediated cancer and cancer stem cells. Int J Mol Sci. (2022) 23. doi: 10.3390/ijms231911746

CrossRef Full Text | Google Scholar

115. Yamada M, Tsukimura N, Ikeda T, Sugita Y, Att W, Kojima N, et al. N-acetyl cysteine as an osteogenesis-enhancing molecule for bone regeneration. Biomaterials. (2013) 34:6147–56. doi: 10.1016/j.biomaterials.2013.04.064

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Chua L. A review on plant-based rutin extraction methods and its pharmacological activities. J Ethnopharmacol. (2013) 150:805–17. doi: 10.1016/j.jep.2013.10.036

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Weidinger A, Milivojev N, Hosmann A, Duvigneau JC, Szabo C, Törö G, et al. Oxoglutarate dehydrogenase complex controls glutamate-mediated neuronal death. Redox Biol. (2023) 62:102669. doi: 10.1016/j.redox.2023.102669

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Moukhadder HM, Halawi R, Cappellini MD, Taher AT. Hepatocellular carcinoma as an emerging morbidity in the thalassemia syndromes: A comprehensive review. Cancer. (2016) 123:751–8. doi: 10.1002/cncr.30462

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Chung W, Lin C, Lin C, Kao C. Thalassaemia and risk of cancer: a population-based cohort study. J Epidemiol Commun H. (2015) 69:1066–70. doi: 10.1136/jech-2014-205075

CrossRef Full Text | Google Scholar

120. Zanella S, Garani M, Borgna-Pignatti C. Malignancies and thalassemia: a review of the literature. Ann NY Acad Sci. (2016) 1368:140–8. doi: 10.1111/nyas.13005

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Origa R, Gianesin B, Longo F, Di Maggio R, Cassinerio E, Gamberini MR, et al. Incidence of cancer and related deaths in hemoglobinopathies: A follow-up of 4631 patients between 1970 and 2021. Cancer. (2022) 129:107–17. doi: 10.1002/cncr.34509

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Pellegrino C, Dragonetti G, Chiusolo P, Rossi M, Orlando N, Teofili L. Acute promyelocytic leukemia in a woman with thalassemia intermedia: case report and review of literature on hematological Malignancies in β-thalassemia patients. Hematol Rep. (2022) 14:310–21. doi: 10.3390/hematolrep14040045

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Karimi M, Giti R, Haghpanah S, Azarkeivan A, Hoofar H, Eslami M. Malignancies in patients with beta-thalassemia major and beta-thalassemia intermedia: a multicenter study in Iran. Pediatr Blood Cancer. (2009) 53:1064–7. doi: 10.1002/pbc.22144

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Hodroj M, Bou-Fakhredin R, Nour-Eldine W, Noureldine H, Noureldine M, Taher A. Thalassemia and Malignancy: An emerging concern? Blood Rev. (2019) 37:100585. doi: 10.1016/j.blre.2019.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Benetatos L, Alymara V, Vassou A, Bourantas K. Malignancies in beta-thalassemia patients: a single-center experience and a concise review of the literature. Int J Lab Hematol. (2008) 30:167–72. doi: 10.1111/j.1751-553X.2007.00929.x

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Steensma D, Gibbons R, Higgs D. Acquired alpha-thalassemia in association with myelodysplastic syndrome and other hematologic Malignancies. BLOOD. (2005) 105:443–52. doi: 10.1182/blood-2004-07-2792

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Ahad Aziz Qureshi P, Razi M, Rehman A, Hassan A. Hodgkin lymphoma with co-existing extramedullary haematopoiesis in a Thalasaemia Major patient: Killing two birds with one stone using PET-CT. J PAK Med Assoc. (2019) 69:1059.

PubMed Abstract | Google Scholar

128. Honig G, Suarez C, Vida L, Lu S, Liu E. Juvenile myelomonocytic leukemia (JMML) with the hematologic phenotype of severe beta thalassemia. Am J Hematol. (1998) 58:67–71. doi: 10.1002/(SICI)1096-8652(199805)58:1<67::AID-AJH12>3.0.CO;2-2

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Mohamed e-S, Ibrahim B, Amr D, Noha e-K, Mokhtar M. Asymmetric dimethylarginine levels in children with β-thalassemia and their correlations to tricuspid regurgitant jet velocity. Pediatr Blood Cancer. (2014) 61:1540–3. doi: 10.1002/pbc.25076

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Tanner M, Galanello R, Dessi C, Smith G, Westwood M, Agus A, et al. A randomized, placebo-controlled, double-blind trial of the effect of combined therapy with deferoxamine and deferiprone on myocardial iron in thalassemia major using cardiovascular magnetic resonance. Circulation. (2007) 115:1876–84. doi: 10.1161/CIRCULATIONAHA.106.648790

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Seyyar S, Tıskaoğlu N, Gürbostan G, Pekpak E, Sayglı O. Increased endothelial cell density in childhood patients with thalassemia major. Eye Contact Lens. (2021) 47:660–3. doi: 10.1097/ICL.0000000000000806

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Levin C, Koren A, Rebibo-Sabbah A, Levin M, Na K, Brenner B, et al. Extracellular vesicle microRNA that are involved in β-thalassemia complications. Int J Mol Sci. (2021) 22. doi: 10.3390/ijms22189760

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Cheung Y, Chan G, Ha S. Arterial stiffness and endothelial function in patients with beta-thalassemia major. Circulation. (2002) 106:2561–6. doi: 10.1161/01.CIR.0000037225.92759.A7

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Kheansaard W, Phongpao K, Paiboonsukwong K, Pattanapanyasat K, Chaichompoo P, Svasti S. Microparticles from β-thalassaemia/HbE patients induce endothelial cell dysfunction. Sci Rep. (2018) 8. doi: 10.1038/s41598-018-31386-6

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Cusmà Piccione M, Piraino B, Zito C, Khandheria B, Di Bella G, De Gregorio C, et al. Early identification of cardiovascular involvement in patients with β-thalassemia major. Am J Cardiol. (2013) 112:1246–51. doi: 10.1016/j.amjcard.2013.05.080

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Wei Q, Boulais P, Zhang D, Pinho S, Tanaka M, Frenette P. Maea expressed by macrophages, but not erythroblasts, maintains postnatal murine bone marrow erythroblastic islands. BLOOD. (2019) 133:1222–32. doi: 10.1182/blood-2018-11-888180

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Jacobsen R, Perkins A, Levesque J. Macrophages and regulation of erythropoiesis. Curr Opin Hematol. (2015) 22:212–9. doi: 10.1097/MOH.0000000000000131

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Angelucci E, Bai H, Centis F, Bafti M, Lucarelli G, Ma L, et al. Enhanced macrophagic attack on beta-thalassemia major erythroid precursors. HAEMATOL-HEMATOL J. (2002) 87:578–83.

Google Scholar

139. Li Z, MacDougald OA. Stem cell factor: the bridge between bone marrow adipocytes and hematopoietic cells. Haematologica. (2019) 104:1689–91. doi: 10.3324/haematol.2019.224188

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Pattira B. Editorial for “Marrow fat content and composition in β-thalassemia: A study using 1 H-MRS. J Magn Reson Imaging. (2021) 53:199–200. doi: 10.1002/jmri.27305

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: beta-thalassemia, osteoblast, impaired hematopoiesis, metabolic abnormalities, ineffective erythropoiesis, microenvironment correction

Citation: Li Z, Yao X, Zhang J, Yang J, Ni J and Wang Y (2024) Exploring the bone marrow micro environment in thalassemia patients: potential therapeutic alternatives. Front. Immunol. 15:1403458. doi: 10.3389/fimmu.2024.1403458

Received: 19 March 2024; Accepted: 22 July 2024;
Published: 05 August 2024.

Edited by:

Anupam Basu, The University of Burdwan, India

Reviewed by:

Kumar Pavanish, Singhealth/Duke-NUS Acedemic Medical Centre, Singapore
Giorgia Moschetti, University of Milan, Italy

Copyright © 2024 Li, Yao, Zhang, Yang, Ni and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yajie Wang, kbb165wyj@sina.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.