Introduction

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin’s lymphoma (NHL), featured by heterogeneity, aggressiveness, and poor prognosis [1, 2]. With the emergence of anti-CD20 monoclonal antibodies, the survival rates for DLBCL patients have improved significantly. However, approximately 40% of cases progress to relapsed/refractory DLBCL [3]. Recently, molecular heterogeneity of DLBCL has been identified as a major factor interfering with clinical treatment responses [4], especially in double-hit lymphoma (DHL) and triple-hit lymphoma (THL) [5, 6]. Investigation of new therapeutic strategies is an urgent need to guide clinical practice.

As a prominent hallmark of cancer, metabolic reprogramming represents the phenotypic outcome and environmental influences on the molecular function of tumor cells [7]. Reorganization of tumor metabolism is characterized by increased assumption of various nutrients, including glutamine [8]. Glutamine is a vital metabolic fuel that is essential for ATP production and precursors biosynthesis [9]. Intracellular glutamine is converted into glutamate and alpha-ketoglutarate (α-KG) to support the tricarboxylic acid (TCA) cycle [10]. Glutamine metabolism also promotes nucleotides and protein biosynthesis to meet the demand for rapid tumor growth [11, 12]. In addition to energy supply, activation of glutamine metabolism contributes to counteracting the adverse effects of ammonia on tumor cells [13]. Therefore, glutamine metabolism plays an important role in tumor growth, microenvironment regulation, and drug resistance [14]. A recent study demonstrated that metabolic reprogramming-mediated glutaminolysis was associated with Bruton’s tyrosine kinase inhibitor (BTKi) resistance in mantle cell lymphoma (MCL) [15], suggesting the promise of targeting metabolism in hematological tumors.

As the major derivative of glutamine, a steady level of α-KG is essential for various biological processes [16, 17], while accumulation of α-KG conversely has tumor-suppressing effects. Elevated level of α-KG suppresses tumorigenesis by switching glucose metabolism [18, 19] and hinders tumor progression by driving tumor cell differentiation [20]. In addition, accumulated α-KG blocks tumor growth by inducing gasdermin C (GSDMC)-mediated pyroptosis [17], indicating that cell death induction by α-KG may be considered a potential strategy for cancer. Consistently, α-KG plays an important role in DLBCL, which depends on the functions of α-KG in promoting TCA process [21]. Since α-KG-induced cell death has yet to be described in DLBCL, investigating the mechanisms of α-KG will hope to provide novel therapeutic strategies for DLBCL patients.

Non-apoptotic cell death is evolving into a new cancer treatment strategy due to its influence on tumor progression and treatment response [22, 23]. As a novel form of non-apoptotic cell death, ferroptosis is derived from iron overload and lipid reactive oxygen species (ROS) accumulation [24]. In contrast to cell apoptosis, ferroptosis has specific morphological features, including shrinkage of mitochondria, reduction of mitochondrial cristae, and condensation of the internal membrane [25,

Fig. 7: A working model of α-KG-induced ferroptosis in DHL.
figure 7

Intracellular α-KG was converted to 2-HG by MDH1, further promoting ROS production. ROS accumulation promoted lipid membrane peroxidation and initiated oxidative DNA damage. TP53 overexpression derived from oxidative DNA damage participated in regulating the ferroptosis pathways. TP53-mediated ferroptosis pathways, together with lipid peroxidation, further led to the generation of ferroptosis.

Metabolic reprogramming is essential for maintaining the biological functions of tumor cells and immune cells [7, 50]. As a kind of aggressive B-cell lymphoma, DLBCL is characterized by metabolic heterogeneity that is associated with drug resistance and immune microenvironment remodeling [51,52,53]. Understanding the metabolic features of DLBCL patients is vital for addressing the unmet medical need. Our work demonstrated an increased glutamine metabolism in DLBCL patients, consistent with the findings that glutamine metabolism is up-regulated to meet the demands of tumorigenesis [54,55,56]. In addition, the critical metabolite of glutamine metabolism, known as glutamine, was associated with adverse outcomes, indicating the prognostic value of glutamine in DLBCL. Given the limitations of single-center samples in representing the disease characteristics, a large sample validation will be conducted in future studies.

The importance of glutamine metabolism in DLBCL refers to its contributions to the TCA cycle, in which the glutamine metabolite α-KG is used as specific fuel [21]. As a highly aggressive lymphoma, α-KG of DLBCL patients was maintained at a low level, indicating an active energy metabolism in DLBCL cells. Since tumor invasiveness was associated with increased serum LDH (ratio>1) [36], serum α-KG was negatively correlated with serum LDH in DLBCL patients. However, α-KG levels showed no significant changes in different LDH-ratio groups, which may be limited by the insufficiency of patient samples. Recent studies demonstrated that the accumulation of intracellular α-KG leads to tumor-suppressing effects [57]. On one hand, α-KG accumulation disrupts glutamine anaplerosis by reversing glutamine-mediated nitrogen metabolism [54, 58]. On the other hand, α-KG is associated with induction of cell death, including pyroptosis and ferroptosis. Accumulative α-KG resulted in oxidative stress, which affected the redox state of cancer cells and ferroptosis induction [59, 60]. Specifically, α-KG induced ferroptosis by altering the redox state in OCI-LY1 and OCI-LY10 cell lines which harbor c-MYC rearrangement/amplification and BCL-2 rearrangement [61]. MYC hyperactivation could enhance the tolerance to oxidative stress and DNA damage, leading to poor prognosis in DHL patients [38]. Interestingly, α-KG-induced ROS significantly disrupted redox homeostasis, which contributed to overcoming MYC-mediated oxidative stress and oxidative DNA damage. Therefore, α-KG might be considered a promising therapeutic strategy for DHL patients.

As an oxidative, iron-dependent form of non-apoptotic cell death, ferroptosis locates at the intersection of metabolic modulation and signaling transduction [62]. Here, we demonstrated that α-KG-induced ferroptosis relied on lipid peroxidation and TP53-mediated signalings. Previous studies illustrate the critical role of TP53 in predicting outcomes of DLBCL patients [63]. Notably, our work highlighted an important mechanism by which TP53 regulated ferroptosis pathways in DHL cells. Interestingly, differentially activated ferroptosis pathways were detected in DLBCL cells with different TP53 mutation states and genetic features. A previous study illustrated that the steady-state levels of TP53 were higher than that of TP53WT OCI-LY10 cells, even though OCI-LY1 cells were featured with mutant TP53 [64]. Given the stable expression of TP53 in OCI-LY1 cells, α-KG-induced ferroptosis might depend on the TP53-mediated reduction of solute carrier family 7 member 11 (SLC7A11) and GPX4 [65]. In contrast to OCI-LY1 cells, ABC-like OCI-LY10 cells constitutively activated the nuclear factor kappa-B (NF-κB) pathway to resist oxidative stress [66, 67]. Notably, NF-κB activation was associated with iron metabolism, indicating the potential of GPX4-independent ferroptosis in OCI-LY10 cells [68, 69]. Future studies will focus on revealing regulatory mechanisms of ferroptosis pathways in different subtypes of DLBCL.

Another vital function of α-KG refers to the induction of DNA damage. Evidence shows cellular DNA damage regulates cell proliferation and death [70, 71]. In particular, the invasive manifestations and chemoresistance of DHL are associated with the hyperactivation of DDR in DHL cells [38]. Accumulation of intracellular α-KG exacerbated oxidative stress and DNA damage to break the protective effects of DDR and eventually induced cell death. Therefore, our results indicate that α-KG treatment may improve the sensitivity of DDR-targeted therapy in DHL, which is expected to be an effective therapeutic option for DHL patients.

In summary, our study demonstrated the importance of glutamine metabolism in the tumorigenesis of DLBCL and the mechanisms of α-KG in ferroptosis induction. α-KG-based therapy may be a promising strategy for DLBCL, particularly for DHL patients.