Abstract
Long non-coding RNA (lncRNA) are emerging as powerful and versatile regulators of transcriptional programs and distinctive biomarkers of progression of T-cell lymphoma. Their role in the aggressive anaplastic lymphoma kinase-negative (ALK–) subtype of anaplastic large cell lymphoma (ALCL) has been elucidated only in part. Starting from our previously identified ALCL-associated lncRNA signature and performing digital gene expression profiling of a retrospective cohort of ALCL, we defined an 11 lncRNA signature able to discriminate among ALCL subtypes. We selected a not previously characterized lncRNA, MTAAT, with preferential expression in ALK– ALCL, for molecular and functional studies. We demonstrated that lncRNA MTAAT contributes to an aberrant mitochondrial turnover restraining mitophagy and promoting cellular proliferation. Functionally, lncRNA MTAAT acts as a repressor of a set of genes related to mitochondrial quality control via chromatin reorganization. Collectively, our work demonstrates the transcriptional role of lncRNA MTAAT in orchestrating a complex transcriptional program sustaining the progression of ALK– ALCL.
Introduction
T-cell lymphoma (TCL) is a complex and heterogeneous group of neoplasms with different biology and outcome.1 Its diagnostic classification is still a challenge, making its treatment suboptimal. Anaplastic lymphoma kinasenegative (ALK–) anaplastic large cell lymphoma (ALCL) is one of the most aggressive subtypes of TCL and is characterized by a dismal prognosis and high mortality.2-4 The molecular details of the pathogenesis of ALK– ALCL are largely unknown, thus limiting the development of targeted strategies.5 Therefore, clarifying the mechanisms underlying this disease is of utmost importance.
The fast and massive implementation of deep sequencing technologies has highlighted the role of the non-coding genome in regulating cancer proliferation, especially in complex tumors without a clear genetic driver.6,7 Particular attention has been paid to a family of transcripts of about 200 bp with no coding potential, known as long non-coding RNA (lncRNA).8 LncRNA possess the striking ability to interact with protein-coding and non-coding transcripts regulating and integrating a complex network of processes simultaneously.9-12 Thanks to their peculiar and versatile features, lncRNA are able to influence – in a context-dependent manner – several aspects of cancer biology, such as cellular proliferation, apoptosis, metabolic reprogramming, genomic instability, drug resistance, invasion, and metastasis.13,14 Thus, deregulating the expression of certain lncRNA can directly influence the pathogenesis and/or progression of different types of cancers, including TCL.15
Previously, we investigated the contribution of lncRNA to ALCL pathogenesis by performing deep transcriptomic profiling of a cohort of ALCL. We identified a unique set of 18 lncRNA overexpressed in neoplastic T-lymphocytes compared to normal T-lymphocytes.16 We were able to show that among those, the lncRNA BlackMamba acts as a major transcriptional regulator of neoplastic T-lymphocytes in the ALK– ALCL subtype, influencing lymphoma progression.16,17 Therefore, the identified lncRNA are not only distinctive biomarkers of disease but also play relevant functional roles in the biogenesis of ALCL.
In this work, we expand our knowledge regarding the functional relevance of lncRNA in the development and progression of ALCL. Starting from our previously identified lncRNA signature and integrating the gene expression profiles of a large cohort of ALCL patients with clinical information, we defined a powerful lncRNA signature that discriminates between ALK– and ALK+ subtypes. We then focused on the XLOC_211989 transcript – which resulted specifically associated with the ALK– ALCL subtype – and characterized its function by using multiple functional approaches. Our data suggest that this non-coding transcript coordinates the expression of a set of genes involved in mitochondrial homeostasis and mitophagy promoting lymphoma cell survival. We named this novel lncRNA MiTophagy and ALK– Anaplastic Lymphoma Associated Transcript (M TA AT ) .
Methods
Patients’ specimens
Fresh and viable cryopreserved cells and formalin-fixed paraffin-embedded (FFPE) sections of two retrospective cohorts of ALCL were isolated from diagnostic/relapsed primary lymphoma biopsies. Diagnoses were assigned according to the World Health Organization classification.1 Tissues used for expression analyses were selected for their high tumor cell content (>50%). The FFPE cohort of samples of ALCL consisted of 54 cases whereas the freshly frozen cohort of samples consisted of 18 ALCL cases. Regarding clinicopathological characteristics of the FFPE cohort eligible for the analysis (n=44), among ALK– ALCL, 17/29 (59%) patients were male and the median age was 70 years (range, 31-88). Regarding ALK+ ALCL, 8/15 (53%) patients were male and the median age was 39 years (range, 21-75). Considering follow-up data available for 29/44 patients, the median follow-up of the cohort was 44 months (range, 2.2-152). The 4-year progression-free survival rate was 70.6% (95% confidence interval: 55.2-90.3). The freshly frozen cohort of ALCL and immunophenotypic features of resting and donor CD4+ T-lymphocytes have been described previously.16
The study was approved through institutional human ethics review boards of the Ethical Committee AVEN and AUSL-IRCCS of Reggio Emilia (287/2018/OSS/IRCCSRE), and patients provided written informed consent in accordance with the Declaration of Helsinki.
RNA extraction and quantitative polymerase chain reaction
Total RNA from cells was extracted by TRIzol (Thermo Fisher Scientific) according to the manufacturer’s instructions. One microgram of total RNA was retrotranscribed using the iScript cDNA kit, (Biorad). The amplified transcript level of each specific gene was normalized on the CHMP2A housekeeping gene. The ΔΔCt quantification method was used for quantitative polymerase chain reaction (RT-qPCR) analyses. The list of primers used is provided in Online Supplementary Table S1.
Antisense LNA GapmeR transfection
MAC2A and TLBR-2 cells (1×106) were transfected with 50 nM Antisense LNA GapmeR for a single knockdown (KD). Antisense LNA GapmeR transfections were performed using the Cell Line Nucleofector Kit SF and Amaxa 4D Nucleofector (program DS-130 for TLBR-2; FI115 for MAC2A). Twenty-four hours after transfection, cells were harvested and plated at 2.5×105 cells/mL. Antisense LNA GapmeR Negative Control (Cat. N/ID: 33951, Qiagen) was used as a negative control. For lncRNA_211989/MTAAT we used two different GapmeRs (Cat. N/ID: 339511, Qiagen); their sequences are provided in Online Supplementary Table S2.
Mitochondrial staining
Mitochondria were stained with Mitotracker (Thermo Fisher Scientific) according to the manufacturer’s instructions. Cells were then harvested, fixed in 4% paraformal-dehyde in phosphate-buffered saline (PBS) 1X for 10 min at room temperature, and spotted on glass slides using Cytospin (Thermo Scientific) as previously reported.18 Dots were washed in PBS 1X three times and nuclei were stained with DAPI.
Tetramethylrhodamine methyl ester perchlorate (TMRM) staining was performed according to the manufacturer’s instructions (Thermo Fisher Scientific), without substantial changes. Cells were then harvested and washed once in RPMI medium without serum. Membrane potential was immediately measured by flow cytometry with a FACSCanto™ II Cell Analyzer (BD Biosciences).
Statistical analysis
Statistical analyses were performed using GraphPad Prism software (GraphPad). Statistical significance was determined using a Student t test. Each experiment was replicated multiple times (>3 up to 6). All analyses were performed using R software version 4.1.3.
Results
XLOC_211989 is a novel biomarker that stratifies patients with ALK– anaplastic large cell lymphoma
We assessed whether the previously generated ALCL-associated lncRNA signature16 might be used to distinguish ALCL subtypes in a retrospective cohort of 54 ALCL cases. Gene expression analyses were performed by digital multiplexing profiling using a custom panel of probes targeting the 17 previously identified lncRNA and four additional genes used for the molecular classification of ALCL subtypes.19
Out of the total of 54 samples, 44 ALCL gene expression profiles passed stringent quality controls and resulted eligible for the analysis (Figure 1A). Of these 44 samples, 15 (34%) were classified as ALK+ ALCL and 29 (66%) scored as ALK– ALCL (Figure 1A, Online Supplementary Figure S1A, B, Online Supplementary Table S4). Focusing on lncRNA, we confirmed that 17/17 (100%) lncRNA were expressed in the ALCL cohort. Principal component analysis showed that lncRNA expression profiles segregated well with ALK+ and ALK– ALCL samples (Figure 1B) resulting in 11 of the 17 (65%) lncRNA significantly deregulated between the two subtypes (Figure 1C, Online Supplementary Figure S1C). In particular: seven lncRNA were overexpressed in ALK– ALCL patients while four lncRNA were overexpressed in ALK+ ALCL patients (Figure 1C). In accordance with our previous observations,16 lncRNA BlackMamba showed a significant, negative correlation with ALK expression (Figure 1C). We focused our attention on the six uncharacterized lncRNA that were significantly associated with the ALK– subtype (Figure 1D). First, we confirmed their association with the ALK– subtype by targeted RT-qPCR performed in a previously published cohort of 15 freshly frozen ALCL.16 From this analysis, XLOC_211989 – herein named lncRNA MTAAT – showed the strongest and most significant association with ALK– ALCL (Figure 1D, E, Online Supplementary Figure S1D). No detectable MTAAT expression was observed in donor resting or activated CD4+ cells (Online Supplementary Figure S1E), further confirming ALCL-restricted MTAAT expression.
To strengthen these observations, we explored the correlation between MTAAT and ALK expression in the retrospective cohort of ALCL samples. Linear regression analysis showed that MTAAT was inversely correlated with ALK expression and positively correlated with lncRNA BlackMamba (Figure 1F). The receiver operating characteristic curve for ALK subtype classification showed that MTAAT has a high capacity (70%) to discriminate between ALK– and ALK+ patients (area under the curve=0.70; 95% confidence interval: 0.54-0.85) (Figure 1G, Online Supplementary Figure S1F).
The MTAAT promoter is bound by RNA polymerase II and enriched for active histone marks
Genomic annotation showed that the MTAAT sequence matches an uncharacterized intergenic transcript encoded on the plus strand of chromosome 3 with an estimated transcript length of 7,189 bp and no predicted alternative isoforms (Figure 2A). In silico analysis predicted four potential open reading frames with irrelevant coding potential within the MTAAT sequence, confirming the non-coding nature of this transcript (Online Supplementary Table S5).
Given the specificity of MTAAT expression in the ALK– ALCL subtype, we sought to define in vitro the molecular mechanisms that control its expression. For this analysis, we chose the two ALK– ALCL cell lines, TLBR-2 and MAC2A, displaying the highest levels of MTAAT expression (Online Supplementary Figure S2A). To identify the promoter of MTAAT, we first analyzed RNA polymerase II (RNAPII) genomic occupancy and histone 3 trimethyl lysine 4 (H3K4me3) profile using chromatin immunoprecipitation (ChIP) followed by sequencing in TLBR-2 cells. A high-density distribution of RNAPII within a 2000 bp region spanning the putative transcription start site (TSS) of MTAAT (P3-P6) was observed. This region was also enriched in H3K4me3, confirming the promoter-like nature of its sequence (Figure 2A). We confirmed the findings by ChIP-qPCR in both TLBR-2 and MAC2A cell lines (Figure 2B, C, Online Supplementary Figure S2B). We also showed that this region is marked by a high level of histone H3 acetyl-lysine 27 (H3K27ac) confirming that this locus is transcriptionally active in these cellular models (Figure 2D, Online Supplementary Figure S2B). Notably, ChIP-qPCR analysis did not show RNAPII or histone modification enrichment in a cell line, CUTLL1, negative for MTAAT expression (Online Supplementary Figure S2C), validating the specificity of our observations. To assess whether the putative promoter of MTAAT is able to transactivate transcription, we cloned the 2000 bp DNA sequence spanning from -1,500 bp to +500 bp of the MTAAT-TSS upstream of a luciferase reporter cassette. In this “promoter-like” configuration, high luciferase activity was detected in both MAC2A and TLBR-2 cells (Figure 2E).
Next, we searched for the signaling pathways underlying MTAAT expression. Based on the genomic profiles observed in ChIP-sequencing, we selected a 500 bp region spanning the TSS of MTAAT and performed a motif search analysis to identify potential transcription factor binding sites. For this, we used the FIMO analysis pipeline20 and identified 117 hypothetical transcription factors (Online Supplementary Table S6). Notably, some transcription factors, including STAT, GATA, and IRF family members, are pertinent to signaling pathways known to be active and deregulated in ALK– ALCL5 (Figure 2F). Specifically, we found a significant enrichment of several pathways related to the cellular response to cytokines, interferon, interleukins, and regulation of T-cell differentiation.
MTAAT is a chromatin-associated long non-coding RNA essential for the transcriptional control of mitochondrial processes
To examine the biological role of MTAAT in TCL, we first studied MTAAT cellular localization performing subcellular fractionation. We found that MTAAT was enriched in the nucleus and strongly associated with the chromatin fraction of lymphoma cells (Figure 3A, Online Supplementary Figure S3A) suggesting a putative role in chromatin organization and gene expression regulation. To investigate the role of this lncRNA in regulating lymphoma transcription, we silenced MTAAT expression by targeting different regions, single or in combination, with gapmer technology. MTAAT expression was measured by RT-qPCR and the delivery of multiple gapmers by electroporation resulted in effective knockdown (KD) (>50%) of MTAAT across all ALK– ALCL cell lines tested (Figure 3B). We then used next-generation RNA sequencing to evaluate the genome-wide transcriptional changes triggered by MTAAT silencing (M TAATKD). TLBR-2 cells were subjected to MTAATKD and RNA was collected 24 h after. In parallel and as a control, a scrambled gapmer was also delivered. RNA-sequencing analysis revealed 2,217 differentially expressed genes in MTAATKD compared to the gapmer control. Among these, 67.5% (1,497 genes) were protein-coding. Specifically, we detected 524 downregulated and 937 upregulated genes upon lncRNA MTAATKD (false discovery rate <0.1) (Figure 3C). These findings suggest a role for MTAAT in both the activation and the repression of transcription. Notably, the genomic regions of MTAAT targets are far beyond chromosome 3 (Figure 3D). This suggests that MTAAT could regulate ALK– ALCL transcriptional programs in trans and at a genome-wide level.
Gene-set enrichment analysis revealed diverse biological processes associated with deregulated genes. Specifically, downregulated transcripts showed significant enrichment in several gene sets related to mitochondrial respiratory chain complexes, DNA damage response, and chromatin organization. In contrast, upregulated transcripts are mostly implicated in immune response, glycolytic process, integrated stress response as well as regulation of mitochondrion organization (Figure 3E). Using RT-qPCR, we validated a representative set of upregulated genes confirming the RNA-sequencing results (Figure 3F).
To exclude off-target effects of the gapmers designed for MTAATKD, we decided to corroborate the results by silencing MTAAT with a CRISPR-interference system, by using doxycycline-inducible dCas9-KRAB and two different single-guide RNA targeting the MTAAT promoter. Following lentiviral transduction of ALK– ALCL cells, we induced dCas9-KRAB for 48 h with doxycycline and evaluated MTAAT expression. RT-qPCR confirmed that both singleguide RNA repressed the level of MTAAT by >60% (Online Supplementary Figure S3B-D). Importantly, the expression of 10/12 (83%) gene targets after dCas9-KRAB-mediated MTAAT silencing was consistent with gapmer MTAATKD, ruling out any off-target effects (Online Supplementary Figure S3E). Collectively, the transcriptional changes that we observed upon MTAATKD indicated that this lncRNA acts as a repressor of a set of genes related to mitochondrial quality control.
MTAAT represses BNIP3 and BNIP3L via histone modifications
To understand how the lncRNA MTAAT regulates the expression of mitochondria-related genes, we evaluated changes in the chromatin organization triggered upon MTAATKD investigating, by ChIP, the distribution of H3K4Me3, H3K27Ac, and RNAPII on BCL2 Interacting Protein 3 (BNIP3) and BCL2 Interacting Protein 3 Like (BNIP3L also known as NIX). These proteins resulted in target genes of lncRNA MTAAT and their loss has been implicated in the accumulation of dysfunctional mitochondria in the hematopoietic system.21,22 After the depletion of MTAAT, H3K4Me3 and H3K27Ac levels increased significantly in BNIP3 and BNIP3L promoters (Figure 4A-C). Likewise, RNA-PII was found to be dramatically enriched around the TSS of both genes upon MTAATKD (Figure 4D). Similar changes were observed in additional MTAAT target genes such as Activating Transcriptional Factor 4 (ATF4) and X-Box Binding Protein 1 (XBP1) (Online Supplementary Figure S4A-D). Concordantly with the gene expression profile, no changes were observed in Optineurin (OPTN) gene (Online Supplementary Figure S4A-E). Furthermore, in silico analysis performed with catRAPID23 showed a high interaction propensity of MTAAT with H3K27 methylation complex (Online Supplementary Figure S4F). Collectively, these data confirm that changes in chromatin markers are directly linked to the activity of MTAAT on its target genes.
To strengthen the clinical relevance of MTAAT regulation on these genes, we investigated the expression of BNIP3 and BNIP3L in the retrospective cohort of ALCL included in this study. In line with the repressive effect of MTAAT, BNIP3 expression was lower in ALK– ALCL than in ALK+ ALCL patients (Figure 4E). A similar gene expression correlation was observed in a panel of non-TCL cell lines (Online Supplementary Figure S4G). In contrast, no significant differences were observed between ALCL subtypes for BNIP3L expression (Figure 4E).
MTAAT sustains the growth of anaplastic large cell lymphoma by regulating mitophagy
The transcriptional changes observed upon MTAATKD are suggestive of specific disruptions in mitochondrial homeostasis, such as an aberrant increase in mitochondrial density or changes in mitochondrial morphology. We wondered whether MTAAT promotes ALCL progression by controlling mitochondrial clearance. First, we investigated whether mitochondrial abundance changes in TCL upon dCas9-KRABinducible MTAATKD. We evaluated mitochondrial mass by Mitotracker staining and cytofluorimetric analysis. This analysis showed a time-dependent reduction in Mitotracker intensity signal upon MTAATKD (Figure 5A), whereas doxycycline treatment alone did not lead to changes (data not shown). Along the same line of evidence, we observed a strong reduction in mitochondrial DNA copy number, as determined by RT-qPCR analysis of the mitochondrial gene ND1 (Figure 5B). Furthermore, the steady-state level of sev eral mitochondrial proteins (cytochrome c oxidase subunit IV, superoxide dismutase, cytochrome, and prohibitin 1), assessed by western blot, confirmed these findings (Figure 5C). Since oxidative function is strictly linked to mitochondrial network dynamics, we evaluated the mitochondrial morphology of Mitotracker-stained cells using immunofluorescence. In a basal condition, ALK– ALCL cells showed the mitochondrial network predominantly distributed around the perinuclear region. Upon depletion of MTAAT, mitochondria displayed a more apical/basal localization which is indicative of a less active mitochondrial state24 (Figure 5D). Concordantly, mitochondrial membrane potential, evaluated by TMRM staining, was reduced upon MTAATKD, suggesting mitochondrial dysfunction (Figure 5E). In line with these data, ALK– ALCL patients with high expression of MTAAT showed a high intensity and diffuse staining for superoxide dismutase compared to those expressing low levels of MTAAT (Online Supplementary Figure S5A).
Growing evidence points to a strong relationship between BNIP3, which acts as an adaptor for tethering mitochondria to nascent autophagosomes, and the activation of a selective form of macroautophagy known as mitophagy.25 Having observed the overexpression of BNIP3 upon MTAATKD, we asked whether the observed changes in mitochondrial mass were due to the activation of mitophagy. First, we assessed whether canonical mitophagy markers can be detected upon MTAAT silencing. Notably, MTAATKD induced a time-dependent decrease of LC3 and of the autophagy receptor SQSTM1/p62 in both cell lines, suggestive of increased autophagy flux. Supporting this, treatment with chloroquine, an autophagy inhibitor that blocks the fusion of autophagosomes with lysosomes, blocked the MTAAT-dependent increase of autophagic flux (Online Supplementary Figure S5B). To strengthen these results, we analyzed the co-localization of specific mitophagy adaptors with autophagosomes. We co-transfected TLBR-2 cells with plasmids encoding LC3-GFP and BNIP3-Flag and analyzed their behavior upon MTAATKD. In control cells, both markers showed diffuse and homogeneous staining across the cytoplasm (Figure 6A). By contrast, upon MTAATKD, both LC3 and BNIP3 accumulated into bright cytoplasmic puncta suggestive of LC3 lipidation and BNIP3 recruitment. Importantly, LC3 and BNIP3 puncta co-localize upon MTAATKD (Figure 6A), indicating active mitophagy.
We hypothesized that tumor cells specifically block mi-tophagy to increase mitochondrial mass and sustain proliferation. We, therefore, asked whether MTAATKD impacts ALK– ALCL cell viability. Notably, growth curve analysis and viability assays showed that depleting MTAAT significantly reduced cellular proliferation in both TLBR-2 and MAC2A cell lines (Figure 6B). No changes were recorded in cell cycle profiles and apoptosis was not induced upon MTAATKD, consistent with energy deprivation, rather than cell death (Online Supplementary Figure S5C, D).
Collectively, our data support a model in which the lncRNA MTAAT exerts its function by stimulating an increase in mi-tochondrial mass – and energy output – which is used by ALK– ALCL cells to sustain cell proliferation (Figure 6C).
Discussion
The implementation of digital gene expression profiling paved the way for application of a transcriptomic approach to the classification of TCL, increasing the precision of diagnosis over conventional methods.26 Progress toward understanding the transcriptional complexity of tumors revealed how coding genes are not the only drivers of cancer progression, with non-coding transcripts, such as lncRNA, regulating essential transcriptional cascades during tumorigenesis.9,27,28 However, how lncRNA drive cellular and clinical phenotypes of aggressive TCL subtypes remains unknown.
In this study, we identified a set of lncRNA that act as molecular classifiers to distinguish ALK+ and ALK– ALCL. We also report the role of one of these lncRNA, which we renamed MTAAT, in regulating mitochondrial turnover and progression of aggressive ALK– ALCL. We identified MTAAT as significantly associated with the ALK– ALCL phenotype in two independent cohorts of ALCL patients: first from a cohort of FFPE diagnostic biopsies analyzed by digital expression profiling with the Nanostring nCounter platform and subsequently in a cohort of frozen tissues by RT-qPCR. We also demonstrated the high accuracy of MTAAT in predicting ALK– subtypes in ALCL classification. Although the use of lncRNA as biomarkers is still in its infancy, our results strongly suggest that digital lncRNA profiling could be integrated into diagnostic panels to improve the accuracy and precision of ALCL stratification.
We selected the lncRNA MTAAT for functional studies based on its association with ALK– ALCL. Analysis of the regulatory elements of MTAAT showed its intrinsic ability to regulate transcription. Transcriptional regulation by lncRNA appears to be a mechanism widely used by hematologic malignancies to control the transcription of selective pathways tuning aberrant proliferation and survival of B and T cells.29-32
By performing RNA sequencing on MTAATKD cell lines, we highlighted how the transcriptional program supported by MTAAT converges on the regulation of mitochondrial pathways. Mechanistic investigations showed that the loss of MTAAT is linked to a unique phenotype characterized by increased mitochondrial turnover through positive mitophagy stimulation, accompanied by a reduction in cell proliferation. Remarkably, lymphomas are characterized as oxidative tumors, indicating a requirement of mitochondrial function for tumor progression.33,34 Mitochondria work as metabolic hubs to support cell growth and proliferation, and act as sensors of intracellular stresses that could threaten survival.35,36 Although the role of mitophagy in lymphoid malignancies is still debated, some evidence indicates that the constitutive repression of autophagy/mitophagy contributes to lymphomagenesis.37- 41 The increase in the mitochondrial pool in tumor cells is also emerging as a key factor in the success of immunotherapeutic treatments,42–44 such as chimeric antigen receptor-expressing T cells.45 These chimeric antigen receptor-expressing cells represent an incredibly promising strategy for the treatment of several malignancies and for this reason, further investigations aimed at elucidating the role of MTAAT are warranted.
Among the downstream targets of MTAAT, we identified BNIP3 whose expression is upregulated upon MTAATKD via chromatin reorganization. BNIP3 was originally reported to function as a BH3-only protein which induced programmed cell death.46,47 More recently, it has been shown to function as a stress-induced mitophagy receptor that interacts directly with LC3 to promote the turnover of otherwise healthy mitochondria.48,49 Although various human solid cancers overexpress BNIP3 as they become hypoxic,50 inactivation of BNIP3 via promoter hypermethylation is a common feature of aggressive and advancedstage cancers such as triple-negative breast cancer, hematologic malignancies and advanced-stage pancreatic cancer.51-54 In these tumors, epigenetic silencing of BNIP3 correlates with high cancer cell proliferation, poor prognostic features, and chemoresistance.55,56 In accordance with this observation, we found a significant reduction of BNIP3 expression in a cohort of ALK– ALCL patients. This finding suggests a tumor-suppressive function of BNIP3 also in the context of ALCL. We speculate that the loss of BNIP3 associated with reduced mitophagy may create a more aggressive tumor phenotype and contribute, at least in part, to the chemoresistance observed in this malignancy. This evidence paves the way for the implementation of targeted therapeutic strategies able to re-express BNIP3 in this lymphoid malignancy.
In conclusion, we have characterized the novel lncRNA MTAAT as a new potential biomarker in the stratification of ALCL patients. Functionally, MTAAT acts as a transcriptional brake on mitophagy, promoting the accumulation of mitochondria and supporting lymphoma progression. These findings corroborate our previous data showing a key role of lncRNA in the control of different transcriptional programs in ALK– ALCL.
Footnotes
- Received December 14, 2022
- Accepted June 20, 2023
Correspondence
Disclosures
No conflicts of interest to disclose.
Contributions
VM and AT performed experiments and analyzed data, BD performed gene expression and Nanostring analyses, VM, FT, ES, and EV performed RNA-sequencing, ChIP-sequencing, and bioinformatics analyses. FR performed FACS analyses and cell sorting experiments, MZ, SA, GI, FS, CF, MP, AR, GM, SL, APDS, and AB provided tissue samples and lymphoma diagnosis. MT and AN provided important experimental and analytic support. AC interpreted the results and helped to discuss the results. VF designed the project, interpreted the results, and wrote the manuscript. All the authors read and approved the final version of the manuscript.
Data-sharing statement
All data generated and/or analyzed in this study are included in this article and its Online Supplementary Appendix. The MTAAT sequence has been deposited in the GenBank database with accession number OM642832. Gene expression profile data are available at the Gene Expression Omnibus (GEO) repository (accession number: GSE217426). RNA-sequencing raw data in fastq.gz format are available in the ArrayExpress repository, dataset E-MTAB-12462 (https://www.ebi.ac.uk/arrayexpress/experiments/E-MTAB-1 2462).
Funding
VM and AT were supported by Fondazione AIRC per la Ricerca sul Cancro (AIRC). FR was supported by Fondazione Umberto Veronesi. VM and AR were supported by Fondazione GRADE Onlus. This study was funded by the Italian Ministry of Health through Ricerca Finalizzata (N. GR-2016-02364298, to VF), Bando per la Valorizzazione della Ricerca Istituzionale 2021- fondi 5 per Mille 2020 (to VF) and Fondazione AIACE (to VF). The study was also partially supported by the Italian Ministry of Health-Ricerca Corrente Annual Program 2024.
Acknowledgments
The authors are grateful to Marina Grassi, Ione Tamagnini, Gloria Venturini, and Riccardo Fuoco for technical help and to all the members of the laboratory for helpful discussion.
References
- Swerdlow SH, Campo E, Pileri SA. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016; 127(20):2375-2390. https://doi.org/10.1182/blood-2016-01-643569PubMedPubMed CentralGoogle Scholar
- Ferreri AJM, Govi S, Pileri SA, Savage KJ. Anaplastic large cell lymphoma, ALK-negative. Crit Rev Oncol Hematol. 2013; 85(2):206-215. https://doi.org/10.1016/j.critrevonc.2012.06.004PubMedGoogle Scholar
- Parrilla Castellar ER, Jaffe ES, Said JW. ALK-negative anaplastic large cell lymphoma is a genetically heterogeneous disease with widely disparate clinical outcomes. Blood. 2014; 124(9):1473-1480. https://doi.org/10.1182/blood-2014-04-571091PubMedPubMed CentralGoogle Scholar
- Savage KJ, Harris NL, Vose JM. ALK- anaplastic large-cell lymphoma is clinically and immunophenotypically different from both ALK+ ALCL and peripheral T-cell lymphoma, not otherwise specified: report from the International Peripheral T-Cell Lymphoma Project. Blood. 2008; 111(12):5496-5504. https://doi.org/10.1182/blood-2008-01-134270PubMedGoogle Scholar
- Fiore D, Cappelli LV, Broccoli A, Zinzani PL, Chan WC, Inghirami G. Peripheral T cell lymphomas: from the bench to the clinic. Nat Rev Cancer. 2020; 20(6):323-342. https://doi.org/10.1038/s41568-020-0247-0PubMedGoogle Scholar
- Rheinbay E, Nielsen MM, Abascal F, for the PCAWG Drivers and Functional Interpretation Working Group. PCAWG Structural Variation Working Group PCAWG Consortium. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes. Nature. 2020; 578(7793):102-111. https://doi.org/10.1038/s41586-020-1965-xPubMedPubMed CentralGoogle Scholar
- van Galen P. Decoding the noncoding cancer genome. Cancer Discov. 2020; 10(5):646-647. https://doi.org/10.1158/2159-8290.CD-20-0285PubMedGoogle Scholar
- Kapranov P, Cheng J, Dike S. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 2007; 316(5830):1484-1488. https://doi.org/10.1126/science.1138341PubMedGoogle Scholar
- Statello L, Guo C-J, Chen L-L, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021; 22(2):96-118. https://doi.org/10.1038/s41580-020-00315-9PubMedPubMed CentralGoogle Scholar
- Marchese FP, Raimondi I, Huarte M. The multidimensional mechanisms of long noncoding RNA function. Genome Biol. 2017; 18(1):206. https://doi.org/10.1186/s13059-017-1348-2PubMedPubMed CentralGoogle Scholar
- Goff LA, Rinn JL. Linking RNA biology to lncRNAs. Genome Res. 2015; 25(10):1456-1465. https://doi.org/10.1101/gr.191122.115PubMedPubMed CentralGoogle Scholar
- Ransohoff JD, Wei Y, Khavari PA. The functions and unique features of long intergenic non-coding RNA. Nat Rev Mol Cell Biol. 2018; 19(3):143-157. https://doi.org/10.1038/nrm.2017.104PubMedPubMed CentralGoogle Scholar
- Huarte M. The emerging role of lncRNAs in cancer. Nat Med. 2015; 21(11):1253-1261. https://doi.org/10.1038/nm.3981PubMedGoogle Scholar
- Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer. 2018; 18(1):5-18. https://doi.org/10.1038/nrc.2017.99PubMedPubMed CentralGoogle Scholar
- Iannello A, Ciarrocchi A, Fragliasso V, Vaisitti T. Lift the curtain on long non-coding RNAs in hematological malignancies: pathogenic elements and potential targets. Cancer Lett. 2022; 536:215645. https://doi.org/10.1016/j.canlet.2022.215645PubMedGoogle Scholar
- Fragliasso V, Verma A, Manzotti G. The novel lncRNA BlackMamba controls the neoplastic phenotype of ALK-anaplastic large cell lymphoma by regulating the DNA helicase HELLS. Leukemia. 2020; 34(11):2964-2980. https://doi.org/10.1038/s41375-020-0754-8PubMedGoogle Scholar
- Tameni A, Sauta E, Mularoni V. The DNA-helicase HELLS drives ALK- ALCL proliferation by the transcriptional control of a cytokinesis-related program. Cell Death Dis. 2021; 12(1):130. https://doi.org/10.1038/s41419-021-03425-0PubMedPubMed CentralGoogle Scholar
- Fragliasso V, Chiodo Y, Ferrari-Amorotti G. Phosphorylation of serine 21 modulates the proliferation inhibitory more than the differentiation inducing effects of C/EBPα in K562 cells. J Cell Biochem. 2012; 113(5):1704-1713. https://doi.org/10.1002/jcb.24040PubMedPubMed CentralGoogle Scholar
- Agnelli L, Mereu E, Pellegrino E. Identification of a 3-gene model as a powerful diagnostic tool for the recognition of ALKnegative anaplastic large-cell lymphoma. Blood. 2012; 120(6):1274-1281. https://doi.org/10.1182/blood-2012-01-405555PubMedGoogle Scholar
- Grant CE, Bailey TL, Noble WS. FIMO: scanning for occurrences of a given motif. Bioinformatics. 2011; 27(7):1017-1018. https://doi.org/10.1093/bioinformatics/btr064PubMedPubMed CentralGoogle Scholar
- Sandoval H, Thiagarajan P, Dasgupta SK. Essential role for Nix in autophagic maturation of erythroid cells. Nature. 2008; 454(7201):232-235. https://doi.org/10.1038/nature07006PubMedPubMed CentralGoogle Scholar
- O’Sullivan TE, Johnson LR, Kang HH, Sun JC. BNIP3- and BNIP3L-mediated mitophagy promotes the generation of natural killer cell memory. Immunity. 2015; 43(2):331-342. https://doi.org/10.1016/j.immuni.2015.07.012PubMedPubMed CentralGoogle Scholar
- Bellucci M, Agostini F, Masin M, Tartaglia GG. Predicting protein associations with long noncoding RNAs. Nat Methods. 2011; 8(6):444-445. https://doi.org/10.1038/nmeth.1611PubMedGoogle Scholar
- Facucho-Oliveira JM, St. John JC. The relationship between pluripotency and mitochondrial DNA proliferation during early embryo development and embryonic stem cell differentiation. Stem Cell Rev Rep. 2009; 5(2):140-158. https://doi.org/10.1007/s12015-009-9058-0PubMedGoogle Scholar
- Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol. 2011; 12(1):9-14. https://doi.org/10.1038/nrm3028PubMedPubMed CentralGoogle Scholar
- Amador C, Bouska A, Wright G. Gene expression signatures for the accurate diagnosis of peripheral T-cell lymphoma entities in the routine clinical practice. J Clin Oncol. 2022; 40(36):4261-4275. https://doi.org/10.1200/JCO.21.02707PubMedPubMed CentralGoogle Scholar
- Palazzo AF, Koonin EV. Functional long non-coding RNAs evolve from junk transcripts. Cell. 2020; 183(5):1151-1161. https://doi.org/10.1016/j.cell.2020.09.047PubMedGoogle Scholar
- Schmitt AM, Chang HY. Long noncoding RNAs in cancer pathways. Cancer Cell. 2016; 29(4):452-463. https://doi.org/10.1016/j.ccell.2016.03.010PubMedPubMed CentralGoogle Scholar
- Doose G, Haake A, Bernhart SH. MINCR is a MYC-induced lncRNA able to modulate MYC’s transcriptional network in Burkitt lymphoma cells. Proc Natl Acad Sci U S A. 2015; 112(38):e5261-5270. Google Scholar
- Zhao P, Ji M-M, Fang Y. A novel lncRNA TCLlnc1 promotes peripheral T cell lymphoma progression through acting as a modular scaffold of HNRNPD and YBX1 complexes. Cell Death Dis. 2021; 12(4):321. https://doi.org/10.1038/s41419-021-03594-yPubMedPubMed CentralGoogle Scholar
- Zhao C-C, Jiao Y, Zhang Y-Y. Lnc SMAD5-AS1 as ceRNA inhibit proliferation of diffuse large B cell lymphoma via Wnt/β-catenin pathway by sponging miR-135b-5p to elevate expression of APC. Cell Death Dis. 2019; 10(4):252. https://doi.org/10.1038/s41419-019-1479-3PubMedPubMed CentralGoogle Scholar
- Cui Y, Xu H, Yang Y. The regulation of miR-320a/XBP1 axis through LINC00963 for endoplasmic reticulum stress and autophagy in diffuse large B-cell lymphoma. Cancer Cell Int. 2021; 21(1):305. https://doi.org/10.1186/s12935-021-01992-yPubMedPubMed CentralGoogle Scholar
- Li M, Teater MR, Hong JY. Translational activation of ATF4 through mitochondrial anaplerotic metabolic pathways is required for DLBCL growth and survival. Blood Cancer Discov. 2022; 3(1):50-65. https://doi.org/10.1158/2643-3230.BCD-20-0183PubMedPubMed CentralGoogle Scholar
- Bhalla K, Jaber S, Nahid MN. Role of hypoxia in diffuse large B-cell lymphoma: metabolic repression and selective translation of HK2 facilitates development of DLBCL. Sci Rep. 2018; 8(1):744. https://doi.org/10.1038/s41598-018-19182-8PubMedPubMed CentralGoogle Scholar
- Giacomello M, Pyakurel A, Glytsou C, Scorrano L. The cell biology of mitochondrial membrane dynamics. Nat Rev Mol Cell Biol. 2020; 21(4):204-224. https://doi.org/10.1038/s41580-020-0210-7PubMedGoogle Scholar
- Eisner V, Picard M, Hajnóczky G. Mitochondrial dynamics in adaptive and maladaptive cellular stress responses. Nat Cell Biol. 2018; 20(7):755-765. https://doi.org/10.1038/s41556-018-0133-0PubMedPubMed CentralGoogle Scholar
- Bertolo C, Roa S, Sagardoy A. LITAF, a BCL6 target gene, regulates autophagy in mature B-cell lymphomas. Br J Haematol. 2013; 162(5):621-630. https://doi.org/10.1111/bjh.12440PubMedPubMed CentralGoogle Scholar
- Wang Z, Xu F, Yuan N. Rapamycin inhibits pre-B acute lymphoblastic leukemia cells by downregulating DNA and RNA polymerases. Leuk Res. 2014; 38(8):940-947. https://doi.org/10.1016/j.leukres.2014.05.009PubMedGoogle Scholar
- Cheng C, Wang T, Song Z. Induction of autophagy and autophagy-dependent apoptosis in diffuse large B-cell lymphoma by a new antimalarial artemisinin derivative, SM1044. Cancer Med. 2018; 7(2):380-396. https://doi.org/10.1002/cam4.1276PubMedPubMed CentralGoogle Scholar
- Nahimana A, Attinger A, Aubry D. The NAD biosynthesis inhibitor APO866 has potent antitumor activity against hematologic malignancies. Blood. 2009; 113(14):3276-3286. https://doi.org/10.1182/blood-2008-08-173369PubMedGoogle Scholar
- Torossian A, Broin N, Frentzel J. Blockade of crizotinib-induced BCL2 elevation in ALK-positive anaplastic large cell lymphoma triggers autophagy associated with cell death. Haematologica. 2019; 104(7):1428-1439. https://doi.org/10.3324/haematol.2017.181966PubMedPubMed CentralGoogle Scholar
- Sainero-Alcolado L, Liaño-Pons J, Ruiz-Pérez MV, Arsenian-Henriksson M. Targeting mitochondrial metabolism for precision medicine in cancer. Cell Death Differ. 2022; 29(7):1304-1317. https://doi.org/10.1038/s41418-022-01022-yPubMedPubMed CentralGoogle Scholar
- Missiroli S, Perrone M, Genovese I, Pinton P, Giorgi C. Cancer metabolism and mitochondria: finding novel mechanisms to fight tumours. eBioMedicine. 2020; 59:102943. https://doi.org/10.1016/j.ebiom.2020.102943PubMedPubMed CentralGoogle Scholar
- Porporato PE, Filigheddu N, Pedro JMB-S, Kroemer G, Galluzzi L. Mitochondrial metabolism and cancer. Cell Res. 2018; 28(3):265-280. https://doi.org/10.1038/cr.2017.155PubMedPubMed CentralGoogle Scholar
- Zhang L, Zhang W, Li Z. Mitochondria dysfunction in CD8+ T cells as an important contributing factor for cancer development and a potential target for cancer treatment: a review. J Exp Clin Cancer Res. 2022; 41(1):227. https://doi.org/10.1186/s13046-022-02439-6PubMedPubMed CentralGoogle Scholar
- Chen G, Ray R, Dubik D. The E1B 19K/Bcl-2–binding protein Nip3 is a dimeric mitochondrial protein that activates apoptosis. J Exp Med. 1997; 186(12):1975-1983. https://doi.org/10.1084/jem.186.12.1975PubMedPubMed CentralGoogle Scholar
- Ray R, Chen G, Vande Velde C. BNIP3 heterodimerizes with Bcl-2/Bcl-XL and induces cell death independent of a Bcl-2 homology 3 (BH3) domain at both mitochondrial and nonmitochondrial sites. J Biol Chem. 2000; 275(2):1439-1448. https://doi.org/10.1074/jbc.275.2.1439PubMedGoogle Scholar
- Hanna RA, Quinsay MN, Orogo AM, Giang K, Rikka S, Gustafsson ÅB. Microtubule-associated protein 1 light chain 3 (LC3) interacts with Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via autophagy. J Biol Chem. 2012; 287(23):19094-19104. https://doi.org/10.1074/jbc.M111.322933PubMedPubMed CentralGoogle Scholar
- Glick D, Zhang W, Beaton M. BNip3 regulates mitochondrial function and lipid metabolism in the liver. Mol Cell Biol. 2012; 32(13):2570-2584. https://doi.org/10.1128/MCB.00167-12PubMedPubMed CentralGoogle Scholar
- Sowter HM, Ferguson M, Pym C. Expression of the cell death genes BNip3 and NIX in ductal carcinoma in situ of the breast; correlation of BNip3 levels with necrosis and grade. J Pathol. 2003; 201(4):573-580. https://doi.org/10.1002/path.1486PubMedGoogle Scholar
- Chourasia AH, Tracy K, Frankenberger C. Mitophagy defects arising from BNip3 loss promote mammary tumor progression to metastasis. EMBO Rep. 2015; 16(9):1145-1163. https://doi.org/10.15252/embr.201540759PubMedPubMed CentralGoogle Scholar
- Koop EA, van Laar T, van Wichen DF, de Weger RA, van der Wall E, van Diest PJ. Expression of BNIP3 in invasive breast cancer: correlations with the hypoxic response and clinicopathological features. BMC Cancer. 2009; 9(1):175. https://doi.org/10.1186/1471-2407-9-175PubMedPubMed CentralGoogle Scholar
- Murai M, Toyota M, Satoh A. Aberrant DNA methylation associated with silencing BNIP3 gene expression in haematopoietic tumours. Br J Cancer. 2005; 92(6):1165-1172. https://doi.org/10.1038/sj.bjc.6602422PubMedPubMed CentralGoogle Scholar
- Okami J, Simeone DM, Logsdon CD. Silencing of the hypoxia-inducible cell death protein BNIP3 in pancreatic cancer. Cancer Res. 2004; 64(15):5338-5346. https://doi.org/10.1158/0008-5472.CAN-04-0089PubMedGoogle Scholar
- Akada M, Crnogorac-Jurcevic T, Lattimore S. Intrinsic chemoresistance to gemcitabine is associated with decreased expression of BNIP3 in pancreatic cancer. Clin Cancer Res. 2005; 11(8):3094-3101. https://doi.org/10.1158/1078-0432.CCR-04-1785PubMedGoogle Scholar
- Erkan M, Kleeff J, Esposito I. Loss of BNIP3 expression is a late event in pancreatic cancer contributing to chemoresistance and worsened prognosis. Oncogene. 2005; 24(27):4421-4432. https://doi.org/10.1038/sj.onc.1208642PubMedGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.