scholarly journals ADAR1 can drive Multiple Myeloma progression by acting both as an RNA editor of specific transcripts and as a DNA mutator of their cognate genes

2020 ◽  
Author(s):  
Rafail Nikolaos Tasakis ◽  
Alessandro Laganà ◽  
Dimitra Stamkopoulou ◽  
David T. Melnekoff ◽  
Pavithra Nedumaran ◽  
...  

ABSTRACTRNA editing is an epitranscriptomic modification of emerging relevance to disease development and manifestations. ADAR1, which resides on human chromosome 1q21, is an RNA editor whose over-expression, either by interferon (IFN) induction or through gene amplification, is associated with increased editing and poor outcomes in Multiple Myeloma (MM). Here we explored the role of ADAR1 in the context of MM progression, by focusing on a group of 23 patients in the MMRF CoMMpass Study for which RNAseq and WES datasets exist for matched pre-and post-relapse samples. Our analysis reveals an acquisition of new DNA mutations on disease progression at specific loci surrounding the sites of ADAR associated (A-to-I) RNA editing. These analyses suggest that the RNA editing enzyme ADAR1 can function as a DNA mutator during Multiple Myeloma (MM) progression, and further imply that guide-targeted RNA editing has the capacity to generate specific mutational signatures at predetermined locations. This dual role of RNA editor and DNA mutator might be shared by other deaminases, such as APOBECs, so that DNA mutation might be the result of collateral damage on the genome by an editing enzyme whose primary job is to re-code the cognate transcript toward specific functional outcomes.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3092-3092
Author(s):  
Rafail Nikolaos Tasakis ◽  
Alessandro Lagana ◽  
Violetta Leshchenko ◽  
David Melnekoff ◽  
Itai Beno ◽  
...  

RNA editing is an epitranscriptomic modification of emerging relevance to disease development and manifestations. Here we identify a novel role of the RNA editing enzyme ADAR1 in multiple myeloma (MM) progression as inducer of cognate DNA mutations. We have previously demonstrated (Lagana et al, ASH 2017) that ADAR1, which resides on human chromosome 1q21, is an RNA editor whose over-expression, either by IFN induction or through gene amplification, is associated with poor outcomes in MM. We now demonstrate robust and reproducible ADAR-mediated RNA editing in MM that increases with disease progression. At the same time, since disease progression is also correlated with the acquisition of new mutations, we asked whether ADAR1 could play the dual role of RNA editor and DNA mutator in MM, especially in the context of relapse. In fact, previous work has revealed that ADAR can exert its functions by acting on DNA/RNA hybrids in vitro (Zheng et al, Nucleic Acids Research 2017), and that DNA mutations at edited sites occur more often than at unedited sites in human and D melanogaster (Popitsch et al, BioRxiv 2017). We performed a careful bioinformatic dissection of matched pre-and post-relapse samples from 21 patients in the MMRF CoMMpass Study. Samples were profiled both with whole-exome sequencing (WES) to identify DNA mutations, and with RNAseq to identify editing instances. WES raw data was processed according to GATK Best Practices to generate alignment files, which were then processed with Samtools to identify mutations. RNAseq data was mapped using the tool GSNAP and processed using REDItools to identify editing events. Downstream analysis revealed a correlation between locations of RNA editing at diagnosis and of DNA mutation at relapse, with regions mutated matching known MM mutational hotspots in genes participating in several pathways that are relevant in MM, such as IFNa, IFNg response, IL2-STAT5 and TNF-NFkB. Finally, we demonstrated that editing at those locations is reproducible in a number of tumor cell lines, and that targeted editing of those locations could also result in the generation of mutations, similar to those we observed from patient data. Overall, we have shown that the RNA editor ADAR1, can also mutate the DNA cognate to the targeted transcript, generating specific mutational signatures at predetermined locations. We further hypothesize that this dual role of RNA editor and DNA mutator might be shared by other deaminases, and we suggest that in some contexts, DNA mutation might be the result of collateral damage on the genome by an editing enzyme whose primary job is to re-code the cognate transcript toward specific functional outcomes. Disclosures Madduri: undation Medicine: Consultancy; Celgene: Consultancy; Abbvie: Consultancy; Takeda: Consultancy. Richter:Adaptive Biotechnologies: Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Speakers Bureau; Bristol-Meyers Squibb: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Karyopharm: Membership on an entity's Board of Directors or advisory committees; Oncopeptides: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees. Chari:Seattle Genetics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Research Funding; Oncoceutics: Research Funding; Novartis Pharmaceuticals: Research Funding; GlaxoSmithKline: Research Funding; Array Biopharma: Research Funding; Karyopharm: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Millennium/Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Consultancy; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Cho:Agenus: Research Funding; Genentech: Honoraria, Research Funding; BMS: Consultancy; GSK: Consultancy; Takeda: Research Funding; Celgene: Honoraria, Research Funding; The Multiple Myeloma Research Foundation: Employment. Jagannath:Celgene: Consultancy; Novartis: Consultancy; Merck: Consultancy; Medicom: Speakers Bureau; Multiple Myeloma Research Foundation: Speakers Bureau; BMS: Consultancy. Parekh:Foundation Medicine Inc.: Consultancy; Karyopharm Inc.: Research Funding; Celgene Corporation: Research Funding.


2021 ◽  
Author(s):  
Junhui Xu ◽  
Liang Gao ◽  
Miao Yan ◽  
Bingjie Wang ◽  
Zhengyang Song ◽  
...  

Abstract Background: Myelomatous pleural effusion (MPE), as a presentation of extramedullary infiltration of multiple myeloma (MM), is rare and associated with poor outcomes without comparatively effective treatment now. The value of the cytokine detection in pleural effusions to MPE has not been reported at present. Case presentation: We herein report a case of refractory and relapsed multiple myeloma which developed bilateral MPE due to disease progression caused by intolerance to various chemotherapy regimens. The cytomorphology and flow cytometry is adopted in the diagnosis confirmation. The chemotherapy containing immunomodulators combined with thoracic catheterization drainage is applied to the patient, showing a certain therapeutic effect. During the course of disease, the changes of cytokine profile in pleural effusion were monitored by Biolegend CBA technology, revealing that the cytokines such as IL-6 and IL-10 related to the tumor load in pleural effusion decreased with the improvement of the disease, while IL-2, IL-4, IL-17A, TNF - α, INF - γ, granzyme A, Granzyme B, perforin and granulysin increased with the improvement of the disease. Conclusions: There is a prospect that the cytokines level in pleural effusion becomes an indication to evaluate treatment response of MPE, and in the light of our finding, immunomodulators, IL-2 and INF - γ may be utilized in treating patients suffering MPE.


2021 ◽  
Vol 53 ◽  
pp. S90
Author(s):  
M.T. Segreto ◽  
D. Di Fusco ◽  
S. Sessa ◽  
G. Di Maggio ◽  
C. Maresca ◽  
...  

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3406-3406
Author(s):  
Elisa Lazzari ◽  
Leslie A Crews ◽  
Maria Anna Zipeto ◽  
Shawn Ali ◽  
Catriona HM Jamieson

Abstract INTRODUCTION Multiple myeloma is a heterogeneous hematopoietic malignancy that represents 10% of all blood cancers and is characterized by malignant plasma cell expansion in the bone marrow, which is rich in pro-inflammatory cytokines such as interleukin-6 (IL-6). The emergence of therapeutically recalcitrant disease even following intensive treatment suggests the existence of a drug-resistant myeloma-initiating population. These myeloma-initiating cells are thought to co-opt stem cell pathways that enhance their capacity to self-renew and become dormant in protective niches. Recently, we and others showed that aberrant RNA editing plays a key role in malignant transformation through activation of the inflammation-responsive RNA editase ADAR1. In human leukemia stem cells, ADAR1 activation is associated with enhanced self-renewal of dormant progenitors, while lentiviral-shRNA ADAR1 knockdown reduces malignant stem cell self-renewal in vivo. Notably, in 30% of multiple myeloma cases copy number amplification of the ADAR locus on chromosome 1q21, which portends a poor prognosis. Thus, the goal of this study was to investigate whether ADAR1-mediated RNA editing in multiple myeloma occurs as a result of chromosome amplification and pro-inflammatory signaling, and to evaluate the effects of prolonged immunomodulatory therapy on ADAR1 activation leading to the emergence of drug-resistant myeloma-initiating cells. METHODS AND RESULTS Increased ADAR1 expression in myeloma patient samples To determine whether gene expression of ADAR1 correlates with chromosome 1q21 amplification, we evaluated existing microarray datasets (Multiple Myeloma Genomics Initiative) from well-characterized patient samples harboring 2 to ≥4 copies of the CKS1B gene, which is located within 500kB adjacent to ADAR as well as the interleukin-6 (IL-6) receptor locus IL6R on chromosome 1q21. Notably, gain of 1q21 copy number is associated with more proliferative disease and poor-risk cytogenetics, which could also be related to ADAR activation. Interestingly, our analysis of ADAR1 gene expression showed increased ADAR1 levels in patients with a CKS1B score of 4 or greater (n=6) compared with patients with a score of 2 (n=18). IL-6 mediated induction of RNA editing activity To evaluate the role of pro-inflammatory cytokine stimulation of ADAR1 in the chromosome 1q21-amplified human myeloma cell line H929, cells were exposed to increasing doses (5-20ng/mL) of recombinant human IL-6. ADAR1 activity, evaluated using a novel diagnostic RNA editing site-specific qPCR (RESSq-PCR) assay to detect cancer stem cell generation, showed that IL-6-treated H929 harbored increased ADAR1 activity. ADAR1 activation after prolonged lenalidomide exposure To investigate the effects of immunomodulatory drug treatment on ADAR1-dependent RNA editing, drug-resistant human myeloma cell lines were experimentally derived by low-dose treatment (1 mM) of H929 cells over 10 weeks. Further incubation with lenalidomide at 10mM selected a robustly drug-resistant population. These cells showed a marked increase in ADAR1 expression and RNA editing activity, as measured by direct sequencing and RESSq-PCR. Long-term lenalidomide treatment of H929 also expanded the CD138-negative fraction, suggesting the emergence of a previously-identified myeloma-initiating cell population. Stromal co-culture of H929 cells with a 1:1 mixture of previously inactivated human bone marrow stromal cell lines (HS-5 and HS-27a) secreting IL-6 and other cytokines also enriched for CD138-negative and CD138-dim populations. CONCLUSION Considering the recently described role for ADAR1 in malignant transformation and cancer stem cell generation, myeloma cells harboring 1q21 amplification could gain a self-renewal advantage through ADAR1 activation. Using RESSq-PCR to detect aberrant RNA editing, we observed increased ADAR1 activity during the evolution of 1q21-amplified human myeloma cells under prolonged drug treatment, coupled with an expansion of the CD138-negative fraction, which was also observed following bone marrow stromal co-culture. Together these data suggest that ADAR1 represents a novel diagnostic and therapeutic target for multiple myeloma. Moreover, this multiple myeloma niche model represents a valuable tool for evaluating novel methods to inhibit aberrant ADAR1 activation in drug-resistant malignancies. Disclosures Jamieson: Sanofi: Honoraria; Roche: Honoraria.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2926-2926
Author(s):  
Antonella Caivano ◽  
Francesco La Rocca ◽  
Alessandra Favole ◽  
Sonia Carturan ◽  
Enrico Bracco ◽  
...  

Abstract Abstract 2926 Introduction Angiogenesis plays a central role in the progression of both solid and hematological tumors. In particular, in multiple myeloma (MM) the critical role of bone marrow (BM) microenvironment and angiogenesis has been well documented. The past decade has witnessed a dramatic improvement in the therapeutic options in MM. However, the disease remains incurable, underscoring the need for continued efforts towards understanding MM biology and exploitation of novel therapeutic approaches. In this setting, monoclonal antibodies against myeloma-specific cell surface antigens represent a promising therapeutic approach, which is however hampered by a lack of appropriate target structures expressed across all pathogenic myeloma cells. The Eph receptors, a large family of receptor tyrosine kinases (RTKs) activated by ephrins binding, have been implicated in many processes involved in malignancy, including alteration of the tumor microenvironment and in angiogenesis, in both of which EpHA3 likely plays an active role. Aberrant expression of EpHA3 is seen in many types of hematolologic malignancies (some leukemic cell lines, T-cell lymphoma, acute lymphoblastic leukemia, myeloproliferative neoplasms) although it is not expressed ubiquitously. Finally, the over-expression of Eph is believed to be sufficient to confer tumorigenic potential although probably further mechanisms can occur to abnormally activate the receptor. Basing on the role of EpHA3 in haematological malignancies, a first-in-class engineered IgG1 antibody targeting the EpHA (KB004) was developed and it is now under phase I clinical trials in USA and Australia for the treatment of EpHA3 overexpressing hematological myeloid malignancies refractory to conventional treatment. We investigated the EpHA3 role and its preferential membrane–bound by GPI linker ligand EFNA5, in MM patients in order to define EpHA3 as new molecular target for a novel therapeutic approach with a specific anti EpHA3 monoclonal antibody. The EpHA3 expression has been studied through a comparative proteomic analysis between BM endothelial cells (ECs) of patients with MM (MMECs) or with monoclonal gammopathy of undetermined significance (MGECs), of control subjects (normal ECs) and in MM cell lines. Methods After written informed consent, BM aspirates have been collected from 20 MM and 4 MGUS patients. Normal ECs were derived from 3 BM aspirates of subjects with anemia due to iron or vitamin B12 deficiency. We analyzed the expression levels of EpHA3 in normal ECs, MGECs and MMECs and MM cell lines evaluating the mRNA and protein levels by RT-qPCR and by WB coupled to ImmunoFluorescence analysis. The biological effects of EpHA3 targeting in MMECs have been studied silencing the EpHA3 mRNA in MMECs and testing them at 72h after silencing in series of functinal assays including viability assay by trypan blue exclusion staining and by in vitro angiogenesis assay followed by measurement of mesh areas and vessel length. Moreover, we studied EFNA5 mRNA expression levels in Normal ECs, MGECs and MMECs and in MM cell lines by PCR. Results Our data showed that EpHA3 mRNA levels are progressively increased from ECs to MGECs reaching the highest values in MMECs. Subsequent analysis by WB and immunofluorescence confirmed EpHA3 protein upregulation among the different EC types. The MMECs in which EpHA3 has been silenced revealed a protein level reduction of approximately 60% when compared to the control. We could not detect major viability defects. Furthermore, in vitro angiogenesis inhibition was marginal when compared to the not silenced counterpart. To know whether EpHA3 may impact not only MM angiogenesis but also plasma cells, three MM cell lines were studied for the EpHA3 expression. We found the plasma cell lines gave constant over expression of EpHA3. Finally, the preliminary data regarding EFNA5 mRNA expression level showed it is expressed in either MMECs and MM plasma cell lines. The evaluation of KB004 effect on MMECs in term of apoptosis induction and in vitro tube formation inhibition, as well as the analysis of EpHA3 levels in primary MM plasma cells are in progress. Conclusions From this study we expect to characterize the role of the EpHA3in MM patients and to provide experimental evidences supporting the possibility of using EpHA3 as a new molecular target for MM by proving the in vitro efficacy of a monoclonal antibody to target the angiogenesis of MM. Disclosures: No relevant conflicts of interest to declare.


2011 ◽  
Vol 64 (1) ◽  
pp. 70-72 ◽  
Author(s):  
Rohit Sharma ◽  
Yujuan Wang ◽  
Pei Zhou ◽  
Richard A. Steinman ◽  
Qingde Wang

Author(s):  
Brian Song ◽  
Yusuke Shiromoto ◽  
Moeko Minakuchi ◽  
Kazuko Nishikura

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Sonia Jimeno ◽  
Rosario Prados-Carvajal ◽  
María Jesús Fernández-Ávila ◽  
Sonia Silva ◽  
Domenico Alessandro Silvestris ◽  
...  

AbstractThe maintenance of genomic stability requires the coordination of multiple cellular tasks upon the appearance of DNA lesions. RNA editing, the post-transcriptional sequence alteration of RNA, has a profound effect on cell homeostasis, but its implication in the response to DNA damage was not previously explored. Here we show that, in response to DNA breaks, an overall change of the Adenosine-to-Inosine RNA editing is observed, a phenomenon we call the RNA Editing DAmage Response (REDAR). REDAR relies on the checkpoint kinase ATR and the recombination factor CtIP. Moreover, depletion of the RNA editing enzyme ADAR2 renders cells hypersensitive to genotoxic agents, increases genomic instability and hampers homologous recombination by impairing DNA resection. Such a role of ADAR2 in DNA repair goes beyond the recoding of specific transcripts, but depends on ADAR2 editing DNA:RNA hybrids to ease their dissolution.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 917-917
Author(s):  
Richard XuFeng ◽  
Qiong Yang ◽  
Youzhong Yuan ◽  
Binfeng Lu ◽  
Tao Cheng ◽  
...  

Abstract Abstract 917 Post-transcriptional regulation such as RNA editing in hematopoiesis and lymphopoiesis is poorly understood. ADAR1 (adenosine deaminase acting on RNA-1) is a RNA editing enzyme essential for embryonic development. Disruption of the ADAR1 gene was shown to cause defective embryonic hematopoiesis (Wang Q et al, Science 2000). Moreover, we have recently obtained direct evidence for the preferential effect of ADAR1 deletion on adult hematopoietic progenitor cells as opposed to the more primitive cells via a RNA-editing dependent mechanism by different conditional gene deletion strategies (Xufeng R et al PNAS 2009, in press). To further determine the role of ADAR1 in T cell development, we generated a mouse model in which ADAR1 was deleted specifically in T lymphocytes by interbreeding ADAR1lox/lox mice with Lck-Cre transgenic mice. In our current study, we report that ADAR1 is essential for T cell differentiation at the late progenitor stage in the thymus, coincident with T cell receptor-α/β expression. In ADAR1lox/loxLck-Cre mice, mature T cells decreased dramatically in peripheral blood, spleen and lymph nodes in comparison to littermate controls. In the thymus, the production of CD4+/CD8+ double positive cells was severely impaired and massive cell death was observed in pre-T cell populations. Within the pro-T cells, ADAR1 deletion resulted in a significant decrease of late progenitor cells but not early progenitor subsets. In both pro-T and pre-T cell stages, defective T cell development preferentially occurred in the beta chain positive cells, but was not apparent in gamma/delta T cells. Our data demonstrated an indispensable role of ADAR1 in early T cell differentiation that correlated with T cell receptor beta chain expression, thereby indicating that RNA editing by ADAR1 is an essential event in T cell development. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 (4) ◽  
Author(s):  
Timothy M. Schmidt ◽  
Rafael Fonseca ◽  
Saad Z. Usmani

AbstractGain of chromosome 1q (+1q) is one of the most common recurrent cytogenetic abnormalities in multiple myeloma (MM), occurring in approximately 40% of newly diagnosed cases. Although it is often considered a poor prognostic marker in MM, +1q has not been uniformly adopted as a high-risk cytogenetic abnormality in guidelines. Controversy exists regarding the importance of copy number, as well as whether +1q is itself a driver of poor outcomes or merely a common passenger genetic abnormality in biologically unstable disease. Although the identification of a clear pathogenic mechanism from +1q remains elusive, many genes at the 1q21 locus have been proposed to cause early progression and resistance to anti-myeloma therapy. The plethora of potential drivers suggests that +1q is not only a causative factor or poor outcomes in MM but may be targetable and/or predictive of response to novel therapies. This review will summarize our current understanding of the pathogenesis of +1q in plasma cell neoplasms, the impact of 1q copy number, identify potential genetic drivers of poor outcomes within this subset, and attempt to clarify its clinical significance and implications for the management of patients with multiple myeloma.


Sign in / Sign up

Export Citation Format

Share Document