Skip to main content
Discover Oncology logoLink to Discover Oncology
. 2023 Jul 18;14:131. doi: 10.1007/s12672-023-00725-z

RIPK1 is aberrantly expressed in multiple B-cell cancers and implicated in the underlying pathogenesis

Baoyu Wu 1,, Jingyu Li 1, Han Wang 1, Jianguo Liu 2, Jiayong Li 2, Fang Sun 2, Dong chuan Feng 2
PMCID: PMC10353973  PMID: 37462822

Abstract

According to the latest epidemiology of the US, B-cell cancers account for > 3% of all new cancer cases and > 80% of non-Hodgkin lymphomas. However, the disease-modifying small molecular drug suitable for most B-cell cancers is still lacking. RIPK1 (receptor-interacting serine/threonine-protein kinase 1) has been observed to be dysregulated and implicated in the pathogenesis of multiple solid cancers, of which, however, the roles in blood cancers are quite unclear. In our study, to identify multi-function targets for B-cell cancer treatment, we reanalyzed a public transcriptomic dataset from the database of Gene Expression Omnibus, which includes CD19+ B-cell populations from 6 normal donors and patients of 5 CLL, 10 FL, and 8 DLBCL. After overlapping three groups (CLL vs. normal, FL vs. normal, and DLBCL vs. normal) of differentially expressed genes (DEGs), we obtained 69 common DEGs, of which 3 were validated by real-time quantitative PCR, including RIPK3, IGSF3, TGFBI. Interestingly, we found that the loss function of RIPK1 significantly increases the proliferation and viability of GM12878 cells (a normal human B lymphocyte cell line). Consistently, overexpression of RIPK1 in TMD8 and U2932 cells effectively inhibited cell proliferation and growth. More importantly, modifying RIPK1 kinase activity by a small molecule (such as necrostain-1, HOIPIN-1, etc.) alters the cell growth status of B-cell lymphoma, showing that RIPK1 exhibits anti-tumor activity in the context of B-cell lymphoma. Taken together, we consider that RIPK1 may be a potential target in the clinical application of B-cell lymphoma (including CLL, DLBCL, and FL) treatment.

Supplementary Information

The online version contains supplementary material available at 10.1007/s12672-023-00725-z.

Keywords: B-cell cancers, RIPK1, Necrostain-1, HOIPIN-1

Introduction

The development of B lymphocytes derived from lymphoid progenitors is a complex cellular phenotypic change and differentiation progress, which involves a web of cell fate decisions in response to internal and external cues drives. During the maturation stage, B-cells with aberration in fate decisions should execute programmed cell death, but rare cells may escape from the quality control system to develop into cancer cells (the so-called B-cell cancers/lymphomas). According to the latest epidemiology of the US, B-cell cancers account for > 3% of all new cancer cases and > 80% of non-Hodgkin lymphomas (the most subcategory of hematological malignancies) [1, 2]. Importantly, the top three B-cell cancers, chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), and diffusion large B-cell lymphoma (DLBCL), account for close to 70% of all non-Hodgkin lymphomas [3]. Although these three lymphomas originate from mature B-cells, they are distinct in the underlying pathogenesis and clinical presentation [1, 3]. Recently, Wong et al. constructed a BAFF (B-cell-activating factor) ligand-based chimeric antigen receptor T cell by targeting three receptors (BAFF-R, BCMA, and TACI), which was demonstrated to be effective in killing multiple B-cell cancers [4]. However, the disease-modifying small molecular drug that is suitable for most B-cell cancers is still lacking.

Receptor-interacting serine/threonine-protein kinase 1(RIPK1), a core component (scaffold function) of (tumor necrosis factor receptor 1) TNFR1 complex, serves as a vital cellular signaling hub that participates in TNF-mediated apoptosis and inflammatory pathways [5, 6]. Also, RIPK1 directly interacts with RIPK3 to promote the activation of necroptosis in a kinase activity-dependent manner [7]. In mouse models, RIPK1 depletion leads to multi-organ inflammation and abnormal cell death in animals [8, 9]. Meanwhile, RIPK1 deficiency in humans also causes severe immunodeficiency, intestinal inflammation, arthritis, etc., showing its evolutionary conserved functions among mammals [10, 11]. In the case of cancers, plenty of studies have recently demonstrated that RIPK1 plays a critical role in cancer immune checkpoint blockade (ICB) by modulating inflammation or immunity-related signaling pathway [1214]. For instance, Cucolo et al. observed the expression level of RIPK1 is strongly associated with interferon-mediated resistance, and genetic deletion of RIPK1 in cancer cells leads to ICB failure by compromising chemokine secretion to decrease ARG1+ suppressive myeloid cells [14]. The vital functions of RIPK1 have been extensively investigated in solid tumors, such as hepatocellular carcinoma, soft-tissue sarcoma, melanoma, cervical Cancer, etc. [7, 1517] However, whether and how RIPK1 is implicated in hematological malignancies (especially B-cell cancers) is still not fully explored.

To identify multi-function targets for B-cell cancer treatment, we reanalyzed a public transcriptomic dataset from the database of Gene Expression Omnibus, which includes CD19+ B-cell populations from 6 normal donors and patients of 5 CCL, 10 FL, and 8 DLBCL. After overlapping three groups (CLL vs. normal, FL vs. normal, and DLBCL vs. normal) of differentially expressed genes (DEGs), we obtained 69 common DEGs, of which 3 were validated using real-time quantitative PCR, including RIPK3, IGSF3, TGFBI, etc. Interestingly, we found that the knockdown of RIPK1 in GM12878 cells (a normal human B lymphocyte cell line) significantly increased cell proliferation and viability. Consistently, overexpression of RIPK1 in TMD8 and U2932 cells effectively inhibited cell proliferation and growth. More importantly, restoration of RIPK1 using HOIPIN-1 treatment significantly inhibited cell proliferation of TMD8 and U2932 cells. Altogether, RIPK1 is a promising target for B-cell lymphoma treatment.

Methods and materials

Human specimen collection and processing

In this study, 17 healthy donors and 55 B-cell lymphoma patients (including 14 children and 41 adults) were enrolled, consisting of 19 chronic lymphocytic leukemia (CLL), 15 diffuse large B-cell patients (DLBCL), and 21 follicular lymphomas (FL). The basic clinical information of patients was summarized in Table 1. The peripheral blood, lymph node, or tonsil tissues were collected (before pharmacological interventions) for the purification of CD19+ B-cells. The written informed consent was obtained from the participants or their families, and all the protocols used in our experiments were approved by the ethics committee of Xuzhou children’s hospital, Xuzhou Medical University.

Table1.

basic clinical information of individuals involved in this study

Variable CLL (n = 19) DLBCL (n = 15) FL (n = 21) Overall (n = 55)
Age at baseline (years)
 Mean (sd) 54.6(20.2) 45.3(27.4) 45.1(24.8) 48.5(24.1)
 Gender [n (%)]
 Male 7(36.8%) 5(33.3%) 9(42.9%) 21(38.2%)
 Female 12(63.2%) 10(66.7%) 12(57.1%) 34(61.8%)
Weight(kg)
 Mean (sd) 58(9.6) 55.3(10.3) 55.4(11.9) 56.3(10.6)
 Height(cm)
 Mean (sd) 159.5(8.2) 155.8(17.2) 152.9(19) 156(15.5)
Ann Arbor Stages
 I 2(10.5%) 5(33.3%) 17(81%) 24(43.6%)
 II 7(36.8%) 7(46.7%) 4(19%) 18(32.7%)
 III 7(36.8%) 2(13.3%) (0%) 9(16.4%)
 IV 2(10.5%) 1(6.7%) (0%) 3(5.5%)
 Missing 1(5.3%) (0%) (0%) 1(1.8%)
ECOG performance status
 0 7(35.6%) 5(58.6%) 9(48%) 21(45.5%)
 1 12(64.4%) 10(41.4%) 12(52%) 34(54.5%)

Stages, classified using the modified Ann Arbor staging system

CLL chronic lymphocytic leukemia, DLBCL diffuse large B cell patients, FL follicular lymphomas

For the purification of CD19+ B-cells, the tissues of the lymph node or tonsil were mechanically dissociated and washed with PBS buffer containing 1% fetal bovine serum (FBS) and 2 mM EDTA. To remove the red blood cells, the grated tissues were incubated with lysis buffer containing 10 mM KHCO3, 150 mM NH4Cl, and 0.12 mM EDTA for 30 min at 4 °C. Then, the CD19+ B-cells were isolated from the treated tissues (consisting of PBMCs) using CD19 MicroBeads from Miltenyi Biotec company (Cat: 130-050-301, Shanghai, China) following the protocol provided by the manufacturer. The obtained CD19+ B-cell was subjected to total RNA isolation.

Cell culture

GM12878 cells were purchased from Coriell Institute for Medical Research (NJ, USA), and cultured in RPMI-1640 containing 2 mM L-glutamine and 15%FBS. OCI-LY3 (ACC 761), RAJI (ACC 319), JEKO-1 (ACC 553), U2932 (ACC 633), WSU-FSCCL(ACC 612), and L428(ACC 197) cells were purchased from DSMZ-German Collection of Microorganisms and Cell Cultures (Braunschweig, GERMANY), and cultured in 85%RPMI-1640 plus 15%FBS. TMD8 (CBP600693) cells were purchased from COBIOER biosciences company (Nanjing China), and cultured in RPMI-1640 containing 10%FBS and 0.05 mM 2-mercaptoethanol. Human embryonic kidney (HEK) 293 T cells used for lentivirus packaging were purchased from American Type Culture Collection and were cultured in Gibco Dulbecco’s Modified Eagle Medium (DMEM) containing 10%FBS. All cells were maintained in a 5% CO2 incubator with an inside 37 °C condition.

Construct stable cell lines

To construct RIPK1 knockdown and overexpression stable cell lines, home-made lentivirus vectors pLKO.1-puro (based on plasmid #8453 backbone from Addgene) and pLVX-M-puro (based on plasmid #125,839 from Addgene) were employed, respectively. For the packaging transgene lentivirus, HEK293T cells were co-transfected with helper plasmids psPAX.2 and pMD2.G with lentivirus backbone vector (carrying RIPK1 coding sequences or shRNA mimics) using Sinofection transfection reagent (STF02, shanghai, China) following manufacturer’s instructions. After 2 days of transfection, lentivirus supernatant was collected and filtered using a 0.45-mm filter. Next, target cells were directly infected using the obtained virus supernatant. After 12 h of infection, the virus-containing medium was replaced with a fresh culture medium and supplied with puromycin (ST551, Beyotime, Shanghai, China) at a dose of 1.0 μg/ml for positive selection. The oligos for the shRNA construct were listed as follows:shRIPK1_#1: Forward, 5ʹ—CCGG CAGC ACAA ATAC GAAC TTC AACT CGAG TTGA AGTT CGTA TTTG TGCT GTTT TTC—3ʹ; Reverse,5ʹ—AATT GAAA AACA GCAC AAAT ACGA ACTT CAAC TCGA GTTG AAGT TCGT ATTT GTGC TG—3ʹ shRIPK1_#2: Forward,5ʹ—CCGG AGGT CATG TTCT TTCA GCTT ACTC GAGT AAGC TGAA AGAA CATG ACCT TTTT TC-3ʹ; Reverse,5ʹ—AATT GAAA AAAG GTCA TGTT CTTT CAGC TTAC TCGA GTAA GCTG AAAG AACA TGAC CT—3ʹ; shRIPK1_#3: Forward,5ʹ—CCGG CCTT GTTG ATAA TGAC TTCC ACTC GAGT GGAA GTCA TTAT CAAC AAGG TTTT TC—3ʹ; Reverse,5ʹ—AATT GAAA AACC TTGT TGAT AATG ACTT CCAC TCGA GTGG AAGT CATT ATCA ACAA GG-3ʹ.

Quantitative real-time PCR (qPCR)

The total RNA of B-cells was isolated using the RNAsimple kit from the Tiangen company (DP419, Beijing, China). Next, cDNA was synthesized from 1 μg total RNA using Quantscript Reversely transcription Kit (KR103, Tiangen, Beijing, China). Finally, qPCR was performed using FastFire qPCR PreMix (Cat: FP207, Tiangen, Beijing, China) on CFX Opus 96 Real-Time PCR System (Bio-rad, USA), and each sample was replicated at least three times. GAPDH served as the internal control, and the relative quantification of genes was calculated based on the ΔΔCt method. The oligos used in the study were listed in Table 2.

Table 2.

Differentially expressed genes (DEGs) identified in this study

ensembl_gene_id baseMean log2FoldChange lfcSE stat pvalue padj symbol description bioType EnterzgeneID HGNC_ID FL120 FL153
OSBPL5 ENSG00000021762 19.25425445 -4.651500455 1.025656934 − 4.535142601 5.76E-06 5.98E-05 OSBPL5 Oxysterol binding protein like 5 [Source:HGNC Symbol;Acc:HGNC:16392] Protein_coding 114,879 HGNC:16,392 9 8
IPCEF1 ENSG00000074706 128.9721346 -4.338224913 0.696358542 − 6.229872477 4.67E-10 1.93E-08 IPCEF1 Interaction protein for cytohesin exchange factors 1 [Source:HGNC Symbol;Acc:HGNC:21204] Protein_coding 26,034 HGNC:21,204 28 61
CST7 ENSG00000077984 35.17745193 -6.351241413 0.993360403 − 6.39369296 1.62E-10 7.71E-09 CST7 Cystatin F [Source:HGNC Symbol;Acc:HGNC:2479] Protein_coding 8530 HGNC:2479 31 25
SMARCD3 ENSG00000082014 106.008591 -4.880376814 0.52035678 − 9.378905008 6.67E-21 4.50E-18 SMARCD3 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily d, member 3 [Source:HGNC Symbol;Acc:HGNC:11108] Protein_coding 6604 HGNC:11,108 85 195
SIGLEC1 ENSG00000088827 20.93313484 -5.631687887 1.204432053 − 4.675803732 2.93E-06 3.41E-05 SIGLEC1 Sialic acid binding Ig like lectin 1 [Source:HGNC Symbol;Acc:HGNC:11127] Protein_coding 6614 HGNC:11,127 2 2
SOX10 ENSG00000100146 16.07498427 -5.687103505 1.001997868 − 5.675764075 1.38E-08 3.69E-07 SOX10 SRY-box transcription factor 10 [Source:HGNC Symbol;Acc:HGNC:11190] Protein_coding 6663 HGNC:11,190 7 22
ADTRP ENSG00000111863 57.63906662 -4.178468198 0.734422715 − 5.689459368 1.27E-08 3.42E-07 ADTRP Androgen dependent TFPI regulating protein [Source:HGNC Symbol;Acc:HGNC:21214] Protein_coding 84,830 HGNC:21,214 108 59
ROPN1B ENSG00000114547 21.90928993 -5.654963228 0.981251932 − 5.763008503 8.26E-09 2.37E-07 ROPN1B Rhophilin associated tail protein 1B [Source:HGNC Symbol;Acc:HGNC:31927] Protein_coding 152,015 HGNC:31,927 23 104
PLCL1 ENSG00000115896 36.2190341 -4.425572473 0.776477649 − 5.699549087 1.20E-08 3.27E-07 PLCL1 P1hospholipase C like 1 (inactive) [Source:HGNC Symbol;Acc:HGNC:9063] Protein_coding 5334 HGNC:9063 9 19
GRIN3B ENSG00000116032 19.29138698 − 4.569998165 0.889077952 − 5.140154643 2.75E− 07 4.71E− 06 GRIN3B Glutamate ionotropic receptor NMDA type subunit 3B [Source:HGNC Symbol;Acc:HGNC:16768] Protein_coding 116,444 HGNC:16,768 7 25
PRRX1 ENSG00000116132 62.90985748 − 7.1904274 1.15242204 − 6.23940462 4.39E− 10 1.83E− 08 PRRX1 Paired related homeobox 1 [Source:HGNC Symbol;Acc:HGNC:9142] Protein_coding 5396 HGNC:9142 13 74
TGFBI ENSG00000120708 70.17753189 − 6.667978561 0.85002716 − 7.844430007 4.35E− 15 7.41E− 13 TGFBI Transforming growth factor beta induced [Source:HGNC Symbol;Acc:HGNC:11771] Protein_coding 7045 HGNC:11,771 59 67
EGR1 ENSG00000120738 3332.677443 5.425843533 0.536016317 10.12253426 4.39E− 24 4.82E− 21 EGR1 Early growth response 1 [Source:HGNC Symbol;Acc:HGNC:3238] protein_coding 1958 HGNC:3238 420 774
PI3 ENSG00000124102 28.54999473 7.326915054 0.870335082 8.418499039 3.81E− 17 1.09E− 14 PI3 Peptidase inhibitor 3 [Source:HGNC Symbol;Acc:HGNC:8947] Protein_coding 5266 HGNC:8947 1 0
FOSB ENSG00000125740 6444.360908 5.409430431 0.710607435 7.612403372 2.69E− 14 3.59E− 12 FOSB FosB proto− oncogene, AP− 1 transcription factor subunit [Source:HGNC Symbol;Acc:HGNC:3797] Protein_coding 2354 HGNC:3797 2990 783
TMEM74B ENSG00000125895 8.682578637 − 5.279355574 0.917646672 − 5.75314632 8.76E− 09 2.48E− 07 TMEM74B Transmembrane protein 74B [Source:HGNC Symbol;Acc:HGNC:15893] Protein_coding 55,321 HGNC:15,893 14 20
GLIS2 ENSG00000126603 19.83998958 − 6.486610596 0.994891921 − 6.519914836 7.03E− 11 3.74E− 09 GLIS2 GLIS family zinc finger 2 [Source:HGNC Symbol;Acc:HGNC:29450] Protein_coding 84,662 HGNC:29,450 12 18
APOE ENSG00000130203 228.4619596 − 6.174274732 0.74239677 − 8.316677794 9.05E− 17 2.35E− 14 APOE Apolipoprotein E [Source:HGNC Symbol;Acc:HGNC:613] protein_coding 348 HGNC:613 63 43
SPINK5 ENSG00000133710 84.04432459 9.16215216 0.863494168 10.61055477 2.66E− 26 5.84E− 23 SPINK5 Serine peptidase inhibitor Kazal type 5 [Source:HGNC Symbol;Acc:HGNC:15464] Protein_coding 11,005 HGNC:15,464 0 0
EMP1 ENSG00000134531 209.6015354 5.383751607 0.620086528 8.682258633 3.88E− 18 1.40E− 15 EMP1 Epithelial membrane protein 1 [Source:HGNC Symbol;Acc:HGNC:3333] Protein_coding 2012 HGNC:3333 5 9
AGT ENSG00000135744 24.58099428 − 7.290746614 0.897751242 − 8.121121169 4.62E− 16 1.03E− 13 AGT Angiotensinogen [Source:HGNC Symbol;Acc:HGNC:333] Protein_coding 183 HGNC:333 25 79
CHRND ENSG00000135902 6.666055491 − 4.897050912 1.038624615 − 4.71493824 2.42E− 06 2.92E− 05 CHRND Cholinergic receptor nicotinic delta subunit [Source:HGNC Symbol;Acc:HGNC:1965] Protein_coding 1144 HGNC:1965 3 8
IL1RN ENSG00000136689 91.37593779 4.836036471 0.712422604 6.788156976 1.14E− 11 7.56E− 10 IL1RN Interleukin 1 receptor antagonist [Source:HGNC Symbol;Acc:HGNC:6000] Protein_coding 3557 HGNC:6000 7 15
IL36A ENSG00000136694 29.47339375 8.772013712 1.242289579 7.061166623 1.65E− 12 1.34E− 10 IL36A interleukin 36 alpha [Source:HGNC Symbol;Acc:HGNC:15562] Protein_coding 27,179 HGNC:15,562 0 0
ENPP2 ENSG00000136960 215.7734377 − 5.943259452 0.926785233 − 6.412768829 1.43E− 10 6.93E− 09 ENPP2 Ectonucleotide pyrophosphatase/phosphodiesterase 2 [Source:HGNC Symbol;Acc:HGNC:3357] Protein_coding 5168 HGNC:3357 20 16
RIPK1 ENSG00000137275 1672.093396 4.512926465 0.4626578 9.754350762 1.77E− 22 1.56E− 19 RIPK1 Receptor interacting serine/threonine kinase 1 [Source:HGNC Symbol;Acc:HGNC:10019] Protein_coding 8737 HGNC:10,019 136 151
EVA1B ENSG00000142694 67.31841674 − 4.542154309 0.835141546 − 5.438783794 5.36E− 08 1.18E− 06 EVA1B Eva− 1 homolog B [Source:HGNC Symbol;Acc:HGNC:25558] Protein_coding 55,194 HGNC:25,558 169 272
IGSF3 ENSG00000143061 128.6702029 − 6.900098858 0.795042876 − 8.678901561 4.00E− 18 1.43E− 15 IGSF3 Immunoglobulin superfamily member 3 [Source:HGNC Symbol;Acc:HGNC:5950] Protein_coding 3321 HGNC:5950 126 200
LCN2 ENSG00000148346 126.7705225 10.30365243 1.003343621 10.26931573 9.69E− 25 1.31E− 21 LCN2 lipocalin 2 [Source:HGNC Symbol;Acc:HGNC:6526] Protein_coding 3934 HGNC:6526 2 0
TCF7L1 ENSG00000152284 20.94621705 − 4.144226314 0.658261951 − 6.295709953 3.06E− 10 1.33E− 08 TCF7L1 transcription factor 7 like 1 [Source:HGNC Symbol;Acc:HGNC:11640] Protein_coding 83,439 HGNC:11,640 25 31
VSIG4 ENSG00000155659 24.52393901 − 5.301813048 0.894588495 − 5.92653838 3.09E− 09 1.00E− 07 VSIG4 V− set and immunoglobulin domain containing 4 [Source:HGNC Symbol;Acc:HGNC:17032] Protein_coding 11,326 HGNC:17,032 4 36
PDZD9 ENSG00000155714 7.852318706 − 5.625245103 0.940083273 − 5.983773207 2.18E− 09 7.38E− 08 PDZD9 PDZ domain containing 9 [Source:HGNC Symbol;Acc:HGNC:28740] Protein_coding 255,762 HGNC:28,740 7 13
C1QC ENSG00000159189 18.49822852 − 5.902796992 1.138299643 − 5.185626675 2.15E− 07 3.82E− 06 C1QC Complement C1q C chain [Source:HGNC Symbol;Acc:HGNC:1245] Protein_coding 714 HGNC:1245 3 5
CHRNB2 ENSG00000160716 73.1995986 − 4.388338218 0.645791563 − 6.795285772 1.08E− 11 7.23E− 10 CHRNB2 Cholinergic receptor nicotinic beta 2 subunit [Source:HGNC Symbol;Acc:HGNC:1962] Protein_coding 1141 HGNC:1962 62 116
GRIN2C ENSG00000161509 23.37444899 − 4.45191811 0.71189064 − 6.253654508 4.01E− 10 1.69E− 08 GRIN2C Glutamate ionotropic receptor NMDA type subunit 2C [Source:HGNC Symbol;Acc:HGNC:4587] Protein_coding 2905 HGNC:4587 46 47
VCAM1 ENSG00000162692 172.2405269 − 10.09135785 1.211972153 − 8.326394152 8.33E− 17 2.20E− 14 VCAM1 Vascular cell adhesion molecule 1 [Source:HGNC Symbol;Acc:HGNC:12663] Protein_coding 7412 HGNC:12,663 14 23
SPRR3 ENSG00000163209 654.3400278 13.25004165 0.959650345 13.80715561 2.31E− 43 8.10E− 39 SPRR3 Small proline rich protein 3 [Source:HGNC Symbol;Acc:HGNC:11268] Protein_coding 6707 HGNC:11,268 1 0
SPRR2D ENSG00000163216 63.48325347 9.594372884 0.952101213 10.07705143 6.98E− 24 7.42E− 21 SPRR2D Small proline rich protein 2D [Source:HGNC Symbol;Acc:HGNC:11264] Protein_coding 6703 HGNC:11,264 0 0
CTLA4 ENSG00000163599 119.3098047 − 4.501004044 0.634188121 − 7.097269556 1.27E− 12 1.07E− 10 CTLA4 Cytotoxic T− lymphocyte associated protein 4 [Source:HGNC Symbol;Acc:HGNC:2505] Protein_coding 1493 HGNC:2505 75 60
RNASE7 ENSG00000165799 33.24187915 8.076245733 0.973362728 8.297262161 1.07E− 16 2.69E− 14 RNASE7 Ribonuclease A family member 7 [Source:HGNC Symbol;Acc:HGNC:19278] Protein_coding 84,659 HGNC:19,278 0 0
C15orf48 ENSG00000166920 63.70796296 5.741708677 0.861358532 6.665875433 2.63E− 11 1.57E− 09 C15orf48 Chromosome 15 open reading frame 48 [Source:HGNC Symbol;Acc:HGNC:29898] Protein_coding 84,419 HGNC:29,898 15 4
CXCL8 ENSG00000169429 50.83518009 4.654179159 0.927750707 5.016626908 5.26E− 07 8.12E− 06 CXCL8 C− X− C motif chemokine ligand 8 [Source:HGNC Symbol;Acc:HGNC:6025] Protein_coding 3576 HGNC:6025 44 1
CRCT1 ENSG00000169509 9.118403483 7.066510361 1.061193459 6.659021782 2.76E− 11 1.63E− 09 CRCT1 Cysteine rich C− terminal 1 [Source:HGNC Symbol;Acc:HGNC:29875] Protein_coding 54,544 HGNC:29,875 1 0
FOS ENSG00000170345 10,877.02658 4.152074154 0.773077814 5.370836003 7.84E− 08 1.61E− 06 FOS Fos proto− oncogene, AP− 1 transcription factor subunit [Source:HGNC Symbol;Acc:HGNC:3796] Protein_coding 2353 HGNC:3796 12,266 2291
KRT78 ENSG00000170423 90.42515389 9.523857489 0.994836317 9.573290925 1.04E− 21 8.08E− 19 KRT78 Keratin 78 [Source:HGNC Symbol;Acc:HGNC:28926] Protein_coding 196,374 HGNC:28,926 1 0
GIMAP8 ENSG00000171115 66.92925053 − 6.195656893 0.942664567 − 6.572493662 4.95E− 11 2.75E− 09 GIMAP8 GTPase, IMAP family member 8 [Source:HGNC Symbol;Acc:HGNC:21792] Protein_coding 155,038 HGNC:21,792 19 28
KRT13 ENSG00000171401 214.4699512 11.06210891 1.003617447 11.02223655 2.99E− 28 1.16E− 24 KRT13 Keratin 13 [Source:HGNC Symbol;Acc:HGNC:6415] Protein_coding 3860 HGNC:6415 0 0
ADCY5 ENSG00000173175 39.95038232 − 4.736502201 0.832071993 − 5.692418731 1.25E− 08 3.38E− 07 ADCY5 Adenylate cyclase 5 [Source:HGNC Symbol;Acc:HGNC:236] Protein_coding 111 HGNC:236 23 17
C1QA ENSG00000173372 31.89538865 − 7.174670906 1.171308337 − 6.125347767 9.05E− 10 3.43E− 08 C1QA Complement C1q A chain [Source:HGNC Symbol;Acc:HGNC:1241] Protein_coding 712 HGNC:1241 5 4
ODF3 ENSG00000177947 16.26498334 − 6.19547321 0.977790294 − 6.336198313 2.36E− 10 1.07E− 08 ODF3 Outer dense fiber of sperm tails 3 [Source:HGNC Symbol;Acc:HGNC:19905] Protein_coding 113,746 HGNC:19,905 12 39
TMPRSS9 ENSG00000178297 15.28991125 − 6.115447494 1.157500743 − 5.28332058 1.27E− 07 2.44E− 06 TMPRSS9 Transmembrane serine protease 9 [Source:HGNC Symbol;Acc:HGNC:30079] Protein_coding 360,200 HGNC:30,079 12 20
SRPK3 ENSG00000184343 27.1006862 − 4.770906899 0.84759279 − 5.628772393 1.81E− 08 4.67E− 07 SRPK3 SRSF protein kinase 3 [Source:HGNC Symbol;Acc:HGNC:11402] Protein_coding 26,576 HGNC:11,402 15 18
TMPRSS11B ENSG00000185873 157.6573824 10.61799964 0.972338439 10.92006571 9.24E− 28 2.95E− 24 TMPRSS11B Transmembrane serine protease 11B [Source:HGNC Symbol;Acc:HGNC:25398] Protein_coding 132,724 HGNC:25,398 0 0
ZACN ENSG00000186919 375.6300258 − 4.405791529 0.555732804 − 7.927895386 2.23E− 15 4.18E− 13 ZACN Zinc activated ion channel [Source:HGNC Symbol;Acc:HGNC:29504] Protein_coding 353,174 HGNC:29,504 788 867
SIGLEC15 ENSG00000197046 19.15500342 − 5.018354002 0.817447971 − 6.139050045 8.30E− 10 3.17E− 08 SIGLEC15 Sialic acid binding Ig like lectin 15 [Source:HGNC Symbol;Acc:HGNC:27596] Protein_coding 284,266 HGNC:27,596 24 37
LYPD2 ENSG00000197353 13.57843739 6.519775246 1.152197754 5.658555767 1.53E− 08 4.03E− 07 LYPD2 LY6/PLAUR domain containing 2 [Source:HGNC Symbol;Acc:HGNC:25215] Protein_coding 137,797 HGNC:25,215 0 0
RNA5S1 ENSG00000199352 236.5579431 5.710386043 0.594844183 9.599801442 8.01E− 22 6.39E− 19 RNA5S1 RNA, 5S ribosomal 1 [Source:HGNC Symbol;Acc:HGNC:34362] rRNA 100,169,751 HGNC:34,362 18 25
SPDYE2 ENSG00000205238 51.23972902 − 4.400601067 0.839793358 − 5.240099872 1.60E− 07 2.98E− 06 SPDYE2 Speedy/RINGO cell cycle regulator family member E2 [Source:HGNC Symbol;Acc:HGNC:33841] Protein_coding 441,273 HGNC:33,841 35 59
SMTNL1 ENSG00000214872 12.78844262 − 6.340824592 1.23594115 − 5.130361256 2.89E− 07 4.91E− 06 SMTNL1 Smoothelin like 1 [Source:HGNC Symbol;Acc:HGNC:32394] Protein_coding 219,537 HGNC:32,394 20 19
CERS1 ENSG00000223802 6.63121223 − 4.358422288 0.975188259 − 4.469313745 7.85E− 06 7.71E− 05 CERS1 Ceramide synthase 1 [Source:HGNC Symbol;Acc:HGNC:14253] Protein_coding 10,715 HGNC:14,253 3 7
CACTIN− AS1 ENSG00000226800 13.34614807 − 5.424277759 0.919272321 − 5.900621214 3.62E− 09 1.15E− 07 CACTIN− AS1 CACTIN antisense RNA 1 [Source:HGNC Symbol;Acc:HGNC:31391] lncRNA 404,665 HGNC:31,391 26 31
EHBP1− AS1 ENSG00000231609 10.45641408 − 4.534220747 0.850447211 − 5.331572244 9.74E− 08 1.95E− 06 EHBP1− AS1 EHBP1 antisense RNA 1 [Source:HGNC Symbol;Acc:HGNC:55766] lncRNA 100,132,215 HGNC:55,766 14 22
SNORD13 ENSG00000239039 22.66583357 4.408655214 0.605682338 7.278824124 3.37E− 13 3.38E− 11 SNORD13 Small nucleolar RNA, C/D box 13 [Source:HGNC Symbol;Acc:HGNC:32711] snoRNA 692,084 HGNC:32,711 7 7
SPRR2A ENSG00000241794 108.697552 10.94894654 0.933412788 11.73001557 8.94E− 32 7.85E− 28 SPRR2A Small proline rich protein 2A [Source:HGNC Symbol;Acc:HGNC:11261] Protein_coding 6700 HGNC:11,261 0 0
LINC00861 ENSG00000245164 153.6992183 − 5.401263923 0.78988884 − 6.838005104 8.03E− 12 5.57E− 10 LINC00861 Long intergenic non− protein coding RNA 861 [Source:HGNC Symbol;Acc:HGNC:45133] lncRNA 100,130,231 HGNC:45,133 17 134
RPL36A− HNRNPH2 ENSG00000257529 119.7391129 5.188862458 0.699613038 7.416760661 1.20E− 13 1.35E− 11 RPL36A− HNRNPH2 RPL36A− HNRNPH2 readthrough [Source:HGNC Symbol;Acc:HGNC:48349] Protein_coding 100,529,097 HGNC:48,349 26 14
NDUFC2− KCTD14 ENSG00000259112 220.1331784 4.812823629 0.619517404 7.768665731 7.93E− 15 1.26E− 12 NDUFC2− KCTD14 NDUFC2− KCTD14 readthrough [Source:HGNC Symbol;Acc:HGNC:42956] Protein_coding 100,532,726 HGNC:42,956 26 48
TEX52 ENSG00000283297 22.02449776 − 4.264995461 0.755919471 − 5.642129389 1.68E− 08 4.35E− 07 TEX52 Testis expressed 52 [Source:HGNC Symbol;Acc:HGNC:53643] Protein_coding 101,929,469 HGNC:53,643 13 30
FL174 FL202 FL238 FL255 FL301 FL303 FL313 FL3A145 TS060911A TS072111A TS072611B TS081111A TS121511A TS121511B
OSBPL5 29 12 27 391 11 6 272 26 2 0 0 2 0 1
IPCEF1 298 104 120 1659 78 46 252 177 10 5 6 7 7 8
CST7 60 10 79 334 65 24 114 45 1 0 0 1 0 1
SMARCD3 245 40 217 588 250 182 259 205 3 4 4 10 2 0
SIGLEC1 82 4 2 139 12 70 36 123 0 0 2 1 0 0
SOX10 21 15 30 97 19 20 91 17 0 0 0 2 0 0
ADTRP 59 44 50 303 54 12 450 79 4 0 0 7 5 4
ROPN1B 50 28 4 31 69 14 58 55 0 0 0 1 0 2
PLCL1 57 37 44 340 25 23 161 53 2 1 0 3 3 2
GRIN3B 42 13 41 163 27 39 43 9 0 0 0 4 0 1
PRRX1 26 34 77 693 81 12 381 28 0 0 1 2 0 0
TGFBI 149 19 51 474 94 184 385 97 0 1 2 0 0 2
EGR1 138 76 157 445 58 118 352 129 8039 3660 8452 491 11,222 8316
PI3 0 0 1 1 1 0 2 0 54 22 12 35 185 16
FOSB 151 96 45 462 176 165 149 130 10,881 9050 17,377 3008 11,987 24,850
TMEM74B 9 8 12 31 15 11 19 39 0 0 0 1 0 0
GLIS2 45 16 28 168 31 20 68 25 0 0 0 1 0 0
APOE 377 82 341 2625 128 160 975 428 4 3 2 3 3 7
SPINK5 1 0 0 1 0 0 2 1 115 26 210 187 365 46
EMP1 14 10 19 42 4 10 35 20 220 31 295 649 862 92
AGT 71 10 74 52 41 49 46 78 0 0 0 0 0 0
CHRND 12 6 13 59 5 4 16 16 0 0 0 1 0 0
IL1RN 1 7 1 2 5 2 55 9 109 73 142 205 360 69
IL36A 0 0 0 0 0 0 1 0 13 1 12 167 74 6
ENPP2 133 72 246 1597 43 22 2602 200 5 3 6 2 7 2
RIPK1 176 396 212 112 123 350 120 129 3720 3743 2798 2330 2684 2843
EVA1B 20 56 94 102 352 62 155 33 3 1 0 12 0 0
IGSF3 335 317 46 135 153 133 776 195 0 2 0 3 2 0
LCN2 0 0 0 0 0 0 1 0 168 13 238 171 817 66
TCF7L1 37 10 48 135 56 24 47 25 1 0 1 2 3 1
VSIG4 96 20 39 72 30 61 30 121 0 1 0 3 0 0
PDZD9 15 9 39 23 10 13 18 11 0 0 0 0 0 0
C1QC 43 5 7 158 7 43 70 80 0 0 0 0 1 1
CHRNB2 186 57 350 165 94 123 138 171 2 2 0 14 0 2
GRIN2C 28 20 29 126 15 30 105 40 0 0 2 4 0 1
VCAM1 99 138 409 1757 139 81 1009 138 0 0 0 0 0 0
SPRR3 0 0 0 0 0 0 0 0 925 149 1015 971 4105 372
SPRR2D 1 0 0 1 0 0 0 0 160 19 38 51 411 69
CTLA4 159 100 168 854 93 59 672 298 5 12 3 6 1 8
RNASE7 1 0 0 1 1 0 0 1 20 4 33 94 174 31
C15orf48 2 1 1 16 0 0 3 0 100 22 117 22 438 51
CXCL8 0 2 0 1 4 0 8 5 30 33 89 51 217 147
CRCT1 0 0 0 0 0 0 0 0 25 2 7 37 19 3
FOS 490 236 113 2534 321 260 1225 369 19,027 13,902 26,445 6251 17,983 39,313
KRT78 1 0 1 0 0 0 1 0 88 10 86 392 295 35
GIMAP8 131 35 56 655 39 19 459 79 1 1 0 2 2 0
KRT13 1 1 0 0 0 0 0 1 256 16 378 671 918 65
ADCY5 46 23 40 413 28 21 249 27 2 1 3 3 1 0
C1QA 119 4 12 250 19 89 99 134 0 0 0 1 0 0
ODF3 29 17 30 127 11 16 29 34 0 0 0 1 0 0
TMPRSS9 52 1 30 175 2 36 19 8 0 0 0 0 1 0
SRPK3 69 28 60 205 27 24 88 31 0 0 0 5 0 1
TMPRSS11B 0 0 1 0 1 0 0 1 160 21 277 440 759 60
ZACN 1029 140 1061 984 595 593 1025 773 6 10 9 62 7 4
SIGLEC15 30 17 53 61 24 27 102 18 0 1 0 2 0 1
LYPD2 0 0 0 3 1 0 0 1 14 3 22 33 75 0
RNA5S1 21 8 2 6 10 50 2 6 399 344 873 51 492 745
SPDYE2 80 83 29 330 36 20 295 63 1 5 0 8 0 0
SMTNL1 47 11 33 75 20 0 50 0 0 0 0 0 0 0
CERS1 7 10 9 32 15 13 22 10 0 0 0 2 0 0
CACTIN-AS1 37 8 16 66 17 14 52 19 0 0 0 2 0 0
EHBP1-AS1 22 15 17 35 15 22 23 18 0 1 0 2 0 0
SNORD13 2 1 2 7 0 1 2 6 101 37 43 4 58 18
SPRR2A 0 0 0 0 0 0 0 0 227 58 64 117 666 133
LINC00861 406 79 113 1910 79 57 322 368 2 1 8 8 1 4
RPL36A-HNRNPH2 13 0 12 0 3 15 17 13 240 255 328 3 246 371
NDUFC2-KCTD14 34 12 13 12 26 25 22 35 544 450 789 0 360 504
TEX52 21 24 58 117 34 39 85 19 0 1 0 5 1 0

Cell proliferation, viability assay

Cell proliferation was determined using Cell Count Kit-8 (CCK-8, Cat: 40203ES60, YEASEN, Shanghai, China). Cell viability was determined using CellQuanti-Blue Cell Viability Assay Kit (CQBL-05 K, BioAssay systems). About 5000 cells (including RIPK1-knockdown cells GM12878/L428 and RIPK1 overexpression cells TMD8/U2932) were seeded in a 96-well microplate containing a culture growth medium for 12 h. Then, cells were subjected to a cell proliferation/viability assay every 12 h (0, 12, 24, 36, and 48 h) following the manufacturer’s instruction. Each sample was detected with at least three replications.

Cell cycle analysis

Propidium Iodide (P1304MP, ThermoFisher Scientific, USA) staining was employed to determine cell cycles. In detail, the collected cells were aliquoted to 1 × 106 cells/100 μL into FACS tubes, followed by twice washing using 1xPBS. Then, cells were centrifuged for 5 min at a speed of 300×g. Furthermore, cells were resuspended using 100 μL of Flow Cytometry Staining Buffer (Cat: # FC001, R&D systems). 10 μL of PI staining solution was added to a control tube of otherwise unstained cells to adjust flow cytometer settings for PI. Cells were mixed gently and incubated for 5 min in the dark. PI fluorescence of samples was determined (using the FL-2 or FL-3 channel) with a FACScan™ instrument. Each detection was repeated at least 3 times. For cell death ratio detection, Annexin V-FITC/PI double-labeled FACS was carried out following the manufacturer’s protocol (Cat: 40302ES20, Yeasen, Shanghai). Flowjo software was used for gate setting FACS original data, and the ggcyto R package (https://bioconductor.org/packages/release/bioc/html/ ggcyto.html) was used for visualization.

Public data analysis

The public high throughput sequencing data were downloaded from the Gene Expression Omnibus database, which includes CD19+ B-cells from primary peripheral blood or lymph node biopsies of normal participants (N = 6), chronic lymphocytic leukemia (CLL, N = 5), follicular lymphoma (FL, N = 10), and diffuse large b cell patients (DLBCL, N = 8).

(GSE145842, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE145842).

The analysis of differentially expressed genes (DEGs) was performed using an R package of DEseq2 (version-1.36, https://bioconductor.org/packages/release/bioc/html/DESeq2.html), with a cutoff of p-adjust < 0.001 and log2 fold changes > 3. The DEGs identified were visualized by volcano plot using an R package of EnhancedVolcano (version-1.14, https://bioconductor.org/packages/release/bioc/html/EnhancedVolcano.html).

Statistical analysis

In this study, data were presented as mean ± (SD), and the replications of all the samples were presented in the figure legends. The software of GraphPad Prism9 (San Diego, USA) was employed for statistical analyses. The student’s t-test was utilized for comparisons between two groups, and a one- or two-way ANOVA test for comparisons between three or more groups. A P value < 0.05 was set as the cutoff of statistical significance.

Results

RIPK1 is down-regulated in tumor cells of lymphoma patients

To identify a common target for B-cell lymphoma in treatment, we analyzed a CD19+ B-cell transcriptomic dataset from the Gene Expression Omnibus (GEO) database, which consists of samples from normal participants (N = 6), chronic lymphocytic leukemia (CLL, N = 5), follicular lymphoma (FL, N = 10), and diffuse large B-cell lymphoma patients (DLBCL, N = 8). Firstly, we identified three groups of differentially expressed genes (DEGs) by comparing CLL, DLBCL, and FL with normal controls, respectively, and then calculated the common DEGs (Fig. 1A and Additional file 1: S1A, B). Fortunately, we obtained 69 common DEGs (Fig. 1B, Table 3). Then, we selected the top 14 high-expressed DEGs and validated the expression in our B-cell lymphoma samples (19 CLL, 21 FL, and 15 DLBCL) using real-time PCR (Fig. 1C). In the pooled samples' real-time PCR, 64.2% (9/14) DEGs were successfully confirmed, such as FOS, RIPK1, SPRR3, APOE, etc. (Fig. 1C). Furthermore, we detected the RNA level of these 9 DEGs in individual samples mentioned above, three of which were further validated, including RIPK1, IGSF3, and TGFB1 (Fig. 1D). In detail, the expression level of RIPK1 was dramatically down-regulated both in the CD19+ B-cells of CLL, DLBCL, and FL when compared with that in normal donors, and in contrast, IGSF3 and TGFB1 expression levels were up-regulated (Fig. 1D). Besides, we estimated the expression of these three genes in 8 lymphocyte lines, including a normal human B lymphocyte cell line (GM12878), and 6 B-cell non-Hodgkin lymphoma (OCI-LY3, JEKO-1, RAJI, TMD8, U2932, WSU-FSCCL) and a Hodgkin's lymphoma cell line (L428). Interestingly, we observed that RIPK1 mRNA levels were decreased in B-cell lymphoma cell lines when compared with GM12878 normal B-cells (Fig. 1E). Taken together, our results indicated that RIPK1 is commonly down-regulated in B-cell lymphoma cells.

Fig. 1.

Fig. 1

RIPK1 is down-regulated in tumor cells of lymphoma patients. A Volcano plot of DEGs of CLL and DLBCL vs. normal donors, respectively. Chronic lymphocytic leukemia (CCL, N = 5) and diffuse large B-cell patients (DLBCL, N = 8) vs. normal donors (N = 6). The cutoff was set at p-adjust < 0.001 and log2 fold change > 3. B Venn gram of three groups of DEGs. CLL and DLBCL vs. normal donor from 1A, FL vs. normal donor from S1B, all the genes in detail were listed in Table 3. C Real-time PCR estimated the mRNA level of the top 14 highly expressed DEGs in B. The CD19+ B-cells of 17 normal donors, 19 CCL, 15 DLBCL, and 21 FL were pooled together, respectively, and then were subjected to real-time PCR. The color shows the mean of the relative expression of each group. N = 3. **P < 0.01 and ***P < 0.001 by one-way ANOVA. D Real-time PCR estimated the mRNA level of 3 DEGs in CD19+ B-cells from the indicated participants. Samples are the same with C. **P < 0.01 and ***P < 0.001 by student’s t-test. E Real-time PCR estimated the mRNA level of RIPK1 in 8 lines of B-cells. Normal human B lymphocyte cell line (GM12878), B-cell non-Hodgkin lymphoma (OCI-LY3, JEKO-1, RAJI, TMD8, U2932, WSU-FSCCL), and a Hodgkin's lymphoma cell line (L428). N = 6, ***P < 0.001 by student’s t-test

Table 3.

Oligos used in this study for real-time PCR

Oligos Forward(5ʹ
—3ʹ)
Reverse (5ʹ—3ʹ)
FOS GGGGCAAGGTGGAACAGTTAT CCGCTTGGAGTGTATCAGTCA
FOSB GCTGCAAGATCCCCTACGAAG ACGAAGAAGTGTACGAAGGGTT
EGR1 CCACGCCGAACACTGACATT GAGGGGTTAGCGAAGGCTG
RIPK1 TGGGCGTCATCATAGAGGAAG CGCCTTTTCCATGTAAGTAGCA
SPRR3 ATGAGTTCTTACCAGCAGAAGC GTTCAGGGACCTTGGTGTAGC
APOE GTTGCTGGTCACATTCCTGG GCAGGTAATCCCAAAAGCGAC
ZACN CCTGGGGTTCAGCATTACCTT GTTGACGTTGAAGAGGGATGG
IGSF3 GCCGAGATTTCATGCTTCACT CGGAACGCTTTCGGGTCATA
RNA5S1 AACGCGCCCGATCTCGTC AGGCGGTCTCCCATCCAAGTA
NDUFC2 ACCCAGAACCCTTACGGTTTC CTCCGCCGGATCAGGTTATC
ADTRP ACCCCAAGGTCCTAGATACTGT AAAGCTGCTAGACCCAAGAGG
TGFBI CTTCGCCCCTAGCAACGAG TGAGGGTCATGCCGTGTTTC
KRT13 GACCGCCACCATTGAAAACAA TCCAGGTCAGTCTTAGACAGAG
ENPP2 TCGCTGTGACAACTTGTGTAAG CCAATGCGACTCTCCTTTGC
GAPDH CTGGGCTACACTGAGCACC AAGTGGTCGTTGAGGGCAATG

Loss-function of RIPK1 contributes to the cell growth of lymphocytes

The aberration of RIPK1 has been extensively established to be implicated in the pathogenesis of multiple categories of solid cancers by altering cell survival, such as pancreatic, colorectal, hepatocellular cancers, etc. [1820]. Therefore, we speculated that the down-regulation of RIPK1 may also play a pathogenic role in B-cell lymphomas. Then, we knock down the expression of RIPK1 in GM12878 (a normal human B lymphocyte cell line) and L428 (a Hodgkin's lymphoma cell line with a relatively high expression of RIPK1) cells using the shRNA vector (Fig. 2A). In the CCK8 assay, we observed that knockdown of RIPK1 in GM12878 cells significantly promoted cell proliferation after 24 h of seeding, and consistently, L428 cells of the shRIPK1 group also dramatically faster than that of the scramble group (Fig. 2B). These results indicate that RIPK1 possesses an inhibitory function in the cell proliferation of B lymphocytes. In the cell viability assay, our results showed that the cell viability of the shRIPK1 group (both in GM12878 and L428 cells) was elevated after 24 h of seeding when compared with that of the scramble group (Fig. 2C). Furthermore, the cell cycle was analyzed using propidium iodide (PI) staining assay. We observed that the loss function of RIPK1 using shRNA in GM12878 cells significantly increased the G2 stage percentage, from 14.3% (in ~ 5000 cells) in the scramble group to 19.8% in the shRNA group (Fig. 2D, E). Consistently, similar results were also found when comparing the shRIPK1 L428 with the scramble one (Fig. 2D, E). These results indicated that RIPK1 possesses an inhibitory function in the proliferative ability of B lymphocytes. Besides, we observed that the loss-function of RIPK1 using shRNA inhibited cell death, evidenced by a reduction of 37.6% and 35.9% of cell death in GM12878 and L428 cells, respectively (Fig. 2F and Additional file 1: S2E). Altogether, the loss function of RIPK1 contributes to the cell growth of lymphocytes.

Fig. 2.

Fig. 2

Loss-function of RIPK1 contributes to the cell growth of lymphocytes A Real-time PCR estimated the RIPK1 knockdown efficiency in GM12878 and L428 cells. Scramble, scramble shRNA without a specific target; shRIPK1, a pool of three independent shRNAs targeting RIPK1. N = 6, ***P < 0.001 by student’s t-test. B CCK8 assay determined the cell growth of GM12878 and L428 cell lines. Cells in A were seeded onto 96-well plates, and 12 h later, cell growth status was detected every 12 h. N = 3, *P < 0.05, **P < 0.01, ***P < 0.001 by two-way ANOVA. C Cell viability of GM12878 and L428 cell lines. Cells in A were seeded onto 96-well plates, and 12 h later, cell viability was detected every 12 h. N = 3, *P < 0.05, **P < 0.01 by two-way ANOVA. DE. PI staining determined the cell cycle distribution of GM12878 and L428 cell lines. ~ 5000 cells cultured for 36 h were collected for PI staining. shRIPK1#1/2/3 indicates independent shRNA targets of RIPK1. Scr, scramble shRNA; N = 6, ***P < 0.001 by student’s t-test. E Annexin V-FITC/PI double-labeled FACS determined the cell death of GM12878 cell lines. Cells in D were used for experiments

Ectopic RIPK1 in lymphocytes suppresses cell growth

To further validate RIPK1’s inhibitory function in B-cell proliferation, we utilized a lentivirus vector to construct RIPK1 stably overexpressed cell lines in TMD8 and U2932 cells, two RIPK1 low-expressed DLBCL cell lines (Fig. 1E). After quantification of RIPK1 using real-time PCR, we found RIPK1 expression was elevated to 9.5-fold and 6.8-fold in TMD8 and U2932, respectively (Fig. 3A). In the CCK8 assay, our results showed that, after 24 h of seeding, the cell growth of RIPK1-overexpressed TMD8 cells was significantly slower than that of the empty vector group (Fig. 3B). Consistently, similar results also were observed in RIPK1-overexpressed U2932 cells (Fig. 3B). These results directly reveal that RIPK1 possesses an inhibitory function in the cell proliferation of B lymphocytes. Meanwhile, we evaluated the cell viability assay for RIPK1-overexpressed TMD8 and U2932 cell lines (Fig. 3C). Our results indicated that the cell viability of RIPK1-overexpressed U2932 cells was decreased after 24 h of seeding, which is similar to that of TMD8 cells (since 36 h) (Fig. 3C). Furthermore, we carried out cell cycle analysis for RIPK1-overexpressed TMD8 and U2932 cell lines using PI staining assay. We observed that ectopic RIPK1 in TMD8 cells significantly decreased the G2 stage percentage, from 19.3% (in ~ 5000 cells) in the empty vector group to 13.4% in the RIPK1 group (Fig. 3D, E). Similarly, we also observed G2 stage percentage decreased from 23.7% to 19.4% in RIPK1 overexpressed U2932 cells. In addition, the cell death ratio was increased by 1.1 and 0.8 folds after overexpression of RIPK1 in U2932 and TMD8 cells, respectively (Fig. 3F and Additional file 1: S3E). Altogether, the loss function of RIPK1 contributes to the cell growth of lymphocytes. Therefore, our findings collectively revealed that RIPK1 possesses inhibitory functions in the proliferative ability of B lymphocytes.

Fig. 3.

Fig. 3

Ectopic RIPK1 in lymphocytes suppresses cell growth A Real-time PCR estimated the RIPK1 knockdown efficiency in TMD8 and U2932 cells. N = 6, ***P < 0.001 by student’s t-test. B CCK8 assay determined cell proliferation of TMD8 and U2932 cell lines. Cells in A were seeded onto 96-well plates, and 12 h later, cell growth status was detected every 12 h. Vector, empty vector; oeRIPK1, RIPK1 overexpression. N = 3, *P < 0.05, **P < 0.01, ***P < 0.001 by two-way ANOVA. C Cell viability of TMD8 and U2932 cell lines. Cells in A were seeded onto 96-well plates, and 12 h later, cell growth status was detected every 12 h. Vector, empty vector; oeRIPK1, RIPK1 overexpression. N = 3, **P < 0.01, ***P < 0.001 by two-way ANOVA. DE. PI staining determined the cell cycle distribution of TMD8 and U2932 cell lines. ~ 5000 cells cultured for 36 h were collected for PI staining. oeRIPK1#1/2/3 indicates three duplications of RIPK1 overexpressed cell lines. Vector, empty vector; oeRIPK1, RIPK1 overexpression. N = 6, *P < 0.05, **P < 0.01, ***P < 0.001 by student’s t-test F. Annexin V-FITC/PI double-labeled FACS determined the cell death of U2932 cell lines. Cells in B were used for experiments.

RIPK1 inhibitor promotes the proliferation of lymphoma cells

A bulk of studies have indicated that RIPK1 has a dual biological role: 1) promoting cell apoptosis and necroptosis in a kinase activity-dependent manner, and 2) promoting cell survival via its scaffolding function in TNFR1 (tumor necrosis factor receptor 1) signaling [21, 22]. Necrostatin-1 (Nec), as the first specific inhibitor of RIPK1, has been extensively demonstrated to possess noticeable effects on inhibiting necroptosis in various diseases, including inflammatory cardiovascular, neurological diseases, etc. [23]. Therefore, we explored the effects of Nec on B-cell lymphoma. In the cell proliferation assay, we observed that Nec treatment (30 μM, based on the previous report [24]) on GM12878/L428 cells significantly promoted cell growth, which was partially blocked by downregulated RIPK1 expression using shRNA (Fig. 4A and Additional file 1: S2A). Meanwhile, cell viability also was increased by Nec treatment on GM12878/L428 cells, and these effects were largely attenuated in the RIPK1 knockdown group (Fig. 4B and Additional file 1: S2B). Furthermore, we observed that Nec treatment elevated G2 percentage both on GM12878 and L428 cells in a dose-dependent manner, and effects were partially abated in RIPK1 knockdown groups (Fig. 4C–D and Additional file 1: S2C). These results indicate that Nec promotes cell proliferation of B-cell lymphoma largely via targeting RIPK1. Besides, we treated GM12878 and L428 cells using another RIPK1 inhibitor, RIPK1-IN-7 (IN-7), which has a low enzymatic IC50 of 11 nM [25]. In our experiments, we found that treatment of IN-7 (5 nM) increased G2 percentage both in GM12878 and L428 cells, which were largely attenuated in RIPK1 knockdown cells (Additional file 1: Fig. S2D). Altogether, inhibition of RIPK1 kinase activity by small molecule inhibitors can efficiently promote cell proliferation.

Fig. 4.

Fig. 4

RIPK1 inhibitor promotes the proliferation of lymphoma cells A CCK8 assay determined cell proliferation of GM12878 lines. Cells were seeded onto 96-well plates and pre-treated with Nec (30 μM) for 24 h, and then, cell growth status was detected by CCK8 assay every 12 h. N = 3, *P < 0.05, **P < 0.01, ***P < 0.001 by two-way ANOVA. B Cell viability of GM12878 cell lines. Cells in A were seeded onto 96-well plates and pre-treated with Nec (30 μM) for 24 h, and then, cell growth status was detected every 12 h. N = 3, *P < 0.05, **P < 0.01, ***P < 0.001 by two-way ANOVA. C-D. PI staining C determined the cell cycle distribution of GM12878 cell lines and the statistical results D. ~ 5000 cells were pre-treated with Nec (30 μM) for 24 h and then collected for PI staining. N = 6, *P < 0.05, ***P < 0.001 by student’s t-test

HOIPIN-1 inhibits the proliferation of lymphoma cells by enhancing RIPK1 function

Due to the down-regulation of RIPK1 expression of kinase activity promoting cell proliferation, we speculated that activation of RIPK1 by small molecules may lead to cell death of B-cell lymphoma. However, to date, there is still no effective and specific RIPK1 agonist available to activate this enzyme in experiments. Numerous previous studies showed that there is a balance between the kinase activity and scaffold function of RIPK1 [9, 26, 27]. Moreover, modulation of RIPK1 scaffold functions via ubiquitination-related enzymes (such as IAP, LUBAC, A20, and CYLD) can largely influence RIPK1-mediated signaling [28]. Therefore, we hypothesized that LUBAC inhibitors, such as HOIPIN-1 (with an IC50 of 2.8 μM [29]) may enhance RIPK1 kinase-mediated cell death by abating its ubiquitination. To this end, we treated TMD8 and U2932 cells with HOIPIN-1 (HOI) and checked the growth status of the cells. Indeed, we observed that HOI treatment (10 μM) on TMD8 and U2932 cells dramatically suppressed cell growth, and these effects were largely attenuated by the RIPK1-knockdown group (Fig. 5A and Additional file 1: S3A). Consistently, cell viability also decreased after HOI treatment on GM12878/L428 cells, which were abated by the knockdown of RIPK1 expression (Fig. 5B and Additional file 1: S3B). In the cell cycle analysis, we observed that HOI treatment decreased G2 percentage both in TMD8 and U2932 cells in a dose-dependent manner (Fig. 5C–D and Additional file 1: S3C–D). More importantly, these effects were effectively attenuated after the knockdown of RIPK1 using shRNA (Fig. 5C–D and Additional file 1: S3C–D). These results indicate that 1) HOI treatment inhibits cell proliferation of B-cell lymphoma largely via targeting RIPK1; 2) restoration of RIPK1 by small molecule inhibitors can efficiently promote cell death of B-cell lymphoma.

Fig. 5.

Fig. 5

HOIPIN-1 inhibits the proliferation of lymphoma cells by blocking RIPK1 function A. CCK8 assay determined cell proliferation of TMD8 lines. Cells were seeded onto 96-well plates and pre-treated with HOIPIN-1 (HOI, 10 μM) for 24 h, and then, cell growth status was detected by CCK8 assay every 12 h. N = 3, *P < 0.05, **P < 0.01, ***P < 0.001 by two-way ANOVA. B Cell viability of TMD8 cell lines. Cells in A were seeded onto 96-well plates and pre-treated with HOI (10 μM) for 24 h, and then, cell viability status was detected every 12 h. N = 3, *P < 0.05, **P < 0.01, ***P < 0.001 by two-way ANOVA. CD. PI staining (C) determined the cell cycle distribution of TMD8 cell lines and the statistical results (D). ~ 5000 cells were pre-treated with HOI at the indicated dose for 24 h and then collected for PI staining. N = 6, *P < 0.05, ***P < 0.001 one-way ANOVA

Discussion and conclusion

A bulk of reports have demonstrated that RIPK1 generally functions as a critical scaffold molecule of the TNFR1 complex, and achieves various inflammatory and pro-survival biological activities by modulating the activation of the MAPK and NF-κB signaling pathways [10, 22]. Meanwhile, as a kinase, RIPK1 physically interacts with RIPK3 to paradoxically promote apoptosis, necroptosis, etc. by catalyzing the phosphorylation of MLKL [3032]. The dual role of RIPK1 in cell survival emphasizes that the unbalance of RIPK1 expression or activity may cause a serious outcome for cells. In our study, we observed that the mRNA level of RIPK1 was significantly down-regulated in the main categories of B-cell lymphoma, including CLL, DLBCL, and FL by re-analyzing the public dataset in the GEO database. Meanwhile, we also validated these results using biopsies we collected by real-time quantitative PCR. These results suggest that characterizing the roles of RIPK1 in B-cell lymphoma may identify a common target for potential therapy of these diseases.

Firstly, we estimated the expression of RIPK1 in 8 lymphocyte lines, including a normal human B lymphocyte cell line (GM12878), and 6 B-cell non-Hodgkin lymphomas (OCI-LY3, JEKO-1, RAJI, TMD8, U2932, WSU-FSCCL) and a Hodgkin's lymphoma cell line (L428). Indeed, we observed that RIPK1 mRNA levels were decreased in 6 B-cell lymphoma cell lines when compared with normal B-cell GM12878. However, RIPK1 in L428 is slightly (but not significantly) lower than that of GM12878 cells. These findings suggest that the down-regulation of RIPK1 may be common in B-cell but not all subtypes of lymphoma cells. However, this conclusion needs more evidence to support. In 2021, Yin et al. reported that RIPK1 depletion occurs in triple-negative breast carcinoma, and the loss function of RIPK1 significantly aggravates the AQP1-driven TNBC progression and metastasis [33]. More recently, Yu et al. demonstrated that SMYD2 contributes to colorectal cancer by targeting RIPK1 and inhibiting the phosphorylation of RIPK1 [34]. Combined with our observations, it is obvious that the aberration of RIPK1 kinase activity contributes to the pathogenesis of multiple cancers, both solid and blood cancers.

To further investigate the roles of RIPK1 in B-cell lymphoma, we knock down the expression of RIPK1 in GM12878 and L428 cells. Interestingly, our results indicate that the knockdown of RIPK1 significantly promotes cell growth and viability, and inhibits cell death both in GM12878 and L428 cells. Meanwhile, we also observed that the G2 percentage is elevated after RIPK1 down-regulation. In contrast, overexpression of RIPK1 in TMD8 and U2932 cells inhibits cell growth and viability and decreases G2 percentage, and promotes cell death. These results support the previously established kinase-dependent pro-apoptosis and -necroptosis functions of RIPK1. Furthermore, we employed two different RIPK1 inhibitors, Necrostatin-1 and RIPK1-IN-7, to confirm that the kinase activity of RIPK1 plays a critical role in the cell growth of normal B lymphocytes and other types of lymphoma cell lines. Our data reveal that inhibition of RIPK1 kinase activity contributes to cell growth of normal B-cells, which is in favor of the RIPK1 knockdown results mentioned above. Noticeably, necrostatin-1, the inhibitor of necroptosis, has been explored to be developed into a small molecule drug for inflammation-, apoptosis- or necroptosis-related diseases, such as cardiovascular diseases, neonatal hypoxia–ischemia, Alzheimer’s disease, Parkinson’s disease, coronavirus disease 2019 (COVID-19), etc. [23, 3537]. According to our evidence, necrostatin-1 (or RIPK1 inhibitors) may increase the risk of B-cell lymphoma occurrence.

On the other hand, we speculated that the RIPK1 agonist may exhibit an effect against the cell growth of B-cell lymphoma. However, no effective and specific RIPK1 agonist is available to date. It has been well established that the ubiquitination of RIPK1 is mainly mediated by E3 ubiquitin ligases LUBAC, by which the kinase activity of RIPK1 is indirectly arrested [38]. To this end, we used HOI to treat TMD8 and U2932 cells. Interestingly, we observed that HOI treatment dramatically suppressed cell growth of TMD8 and U2932, and increased the G1 arrest of these two cell lines. These findings reveal that restoration of RIPK1 by small molecule inhibitors can efficiently promote cell death of B-cell lymphoma. Notably, HOI is well-known as an inhibitor of the canonical NF-kB by suppressing LUBAC ubiquitin ligase activity [39]. In our recent results (data not shown), we also observed that the p65 phosphorylation (the activated NF-kB subunit) level was significantly decreased by HOI treatment in both TMD8 and U2932 cells. Meanwhile, we found overexpression of RIPK1 inhibited NF-kB activation, which is similar to HOI treatment. These results suggest that HOI is versatile in suppressing cell apoptosis, via both NF-kB and RIPK1-dependent mechanisms. Besides, considering the dramatic inhibitory effect of RIPK1 specific inhibitor on cell death, we hold that RIPK1 promotes cell death by both kinase- and scaffold-mediated function.

In summary, our study reveals that RIPK1 is down-regulated in B-cell lymphoma, including CLL, DLBCL, and FL. Meanwhile, RIPK1 exhibits anti-tumor activity in a kinase-dependent manner in the context of B-cell lymphoma. More importantly, modifying RIPK1 kinase activity by a small molecule (such as necrostain-1, HOIPIN-1, etc.) alters the cell growth status of B-cell lymphoma. Taken together, we consider that RIPK1 may be a potential target in the clinical application of B-cell lymphoma treatment.

Supplementary Information

12672_2023_725_MOESM1_ESM.pdf (1MB, pdf)

Additional file 1: Fig S1. RIPK1 is down-regulated in tumor cells of lymphoma patients. A Heatmap of 3 groups of DEGs (CLL, DLBCL, and FL vs. normal, respectively). Chronic lymphocytic leukemia (CCL, N=5), follicular lymphoma (FL, N=10), and diffuse large B-cell patients (DLBCL, N=8) vs. normal donors (N=6). The cutoff was set at p-adjust < 0.001 and log2 fold change > 3. B Volcano plot of DEGs of FL vs. normal. Follicular lymphoma (FL, N=10) vs. normal donors (N=6). A cutoff of p-adjust < 0.001 and log2 fold change > 3. Fig S2. RIPK1 inhibitor promotes the proliferation of lymphoma cells A CCK8 assay determined cell proliferation of L428 lines. Cells were seeded onto 96-well plates and pre-treated with Nec (30 μM) for 24 h, and then, cell growth status was detected by CCK8 assay every 12 h. N=3, *P<0.05, **P<0.01, ***P<0.001 by two-way ANOVA. B Cell viability of L428 cell lines. Cells in A were seeded onto 96-well plates and pre-treated with Nec (30 μM) for 24 h, and then, cell growth status was detected every 12 h. N=3, *P<0.05, ***P<0.001 by two-way ANOVA. C PI staining determined the cell cycle distribution of L428 cell lines and the statistical results. ~5000 cells were pre-treated with Nec (30 μM) for 24 h and then collected for PI staining. D PI staining determined the cell cycle distribution of GM12878/L428 cell lines and the statistical results. ~5000 cells were pre-treated with RIPK1-IN-7 (IN-7, 5 nM) for 24 h and then collected for PI staining. N=6, ***P<0.001 by student’s t-test. E Annexin V-FITC/PI double-labeled FACS determined the cell death of L428 cell lines. Cells in A were used for experiments. Fig S3. HOIPIN-1 inhibits the proliferation of lymphoma cells by blocking RIPK1 function. A CCK8 assay determined cell proliferation of U2932 lines. Cells were seeded onto 96-well plates and pre-treated with HOIPIN-1 (HOI, 10 μM) for 24 h, and then, cell growth status was detected by CCK8 assay every 12 h. N=3, *P<0.05, **P<0.01, ***P<0.001 by two-way ANOVA. B Cell viability of U2932 cell lines. Cells in A were seeded onto 96-well plates and pre-treated with HOI (10 μM) for 24 h, and then, cell growth status was detected every 12 h. N=3, *P<0.05, ***P<0.001 by two-way ANOVA. CD. PI staining determined the cell cycle distribution of U2932 cell lines and the statistical results D. ~5000 cells were pre-treated with HOI at the indicated dose for 24 h and then collected for PI staining. N=6, *P<0.05, ***P<0.001 by one-way ANOVA.E. Annexin V-FITC/PI double-labeled FACS determined the cell death of TMD8 cell lines. Cells in Fig. 3B were used for experiments.

Acknowledgements

We thank the individuals and their families who participated in this project. This study was funded by Grants from the Applied Basic Research Program of Xuzhou (Nos. KC19031). The funders had no role in study design, data collection and analysis, manuscript preparation, or decision to publish.

Author contributions

BW conceived of this study and obtained financial support. BW wrote and revised the manuscript, JL, HW, JL, JL, FS, and DF performed research and analyzed data.

Funding

This work was supported by Grants from the Applied Basic Research Program of Xuzhou (Nos. KC19031).

Declarations

Competing interests

The authors declare no competing interests.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Armitage JO, et al. Non-HODGKIN LYMPHOMA. The Lancet. 2017;390(10091):298–310. doi: 10.1016/S0140-6736(16)32407-2. [DOI] [PubMed] [Google Scholar]
  • 2.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30. doi: 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
  • 3.Hanel W, Epperla N. Evolving therapeutic landscape in follicular lymphoma: a look at emerging and investigational therapies. J Hematol Oncol. 2021;14(1):104. doi: 10.1186/s13045-021-01113-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wong DP, et al. A BAFF ligand-based CAR-T cell targeting three receptors and multiple B cell cancers. Nat Commun. 2022;13(1):217. doi: 10.1038/s41467-021-27853-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ashida H, Sasakawa C, Suzuki T. A unique bacterial tactic to circumvent the cell death crosstalk induced by blockade of caspase-8. EMBO J. 2020;39(17):e104469. doi: 10.15252/embj.2020104469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Tao P, et al. A dominant autoinflammatory disease caused by non-cleavable variants of RIPK1. Nature. 2020;577(7788):109–114. doi: 10.1038/s41586-019-1830-y. [DOI] [PubMed] [Google Scholar]
  • 7.Yamamoto S, Iwakuma T. RIPK1-TRAF2 interplay on the TNF/NF-κB signaling, cell death, and cancer development in the liver. Transl Cancer Res. 2017;6(Suppl 3):94–109. doi: 10.21037/tcr.2017.04.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Vanlangenakker N, et al. cIAP1 and TAK1 protect cells from TNF-induced necrosis by preventing RIP1/RIP3-dependent reactive oxygen species production. Cell Death Differ. 2011;18(4):656–665. doi: 10.1038/cdd.2010.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Newton K. Multitasking kinase RIPK1 regulates cell death and inflammation. Cold Spring Harb Perspect Biol. 2020 doi: 10.1101/cshperspect.a036368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Takahashi N, et al. RIPK1 ensures intestinal homeostasis by protecting the epithelium against apoptosis. Nature. 2014;513(7516):95–99. doi: 10.1038/nature13706. [DOI] [PubMed] [Google Scholar]
  • 11.Shutinoski B, et al. K45A mutation of RIPK1 results in poor necroptosis and cytokine signaling in macrophages, which impacts inflammatory responses in vivo. Cell Death Differ. 2016;23(10):1628–1637. doi: 10.1038/cdd.2016.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Vredevoogd DW, et al. Augmenting immunotherapy impact by lowering tumor TNF cytotoxicity threshold. Cell. 2019;178(3):585–599. doi: 10.1016/j.cell.2019.06.014. [DOI] [PubMed] [Google Scholar]
  • 13.Sriram G, et al. The injury response to DNA damage in live tumor cells promotes antitumor immunity. Sci Signal. 2021;14(705):eabc4764. doi: 10.1126/scisignal.abc4764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cucolo L, et al. The interferon-stimulated gene RIPK1 regulates cancer cell intrinsic and extrinsic resistance to immune checkpoint blockade. Immunity. 2022;55(4):671–685.e10. doi: 10.1016/j.immuni.2022.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Smith HG, et al. RIPK1-mediated immunogenic cell death promotes anti-tumour immunity against soft-tissue sarcoma. EMBO Mol Med. 2020;12(6):e10979. doi: 10.15252/emmm.201910979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Luan Q, et al. RIPK1 regulates survival of human melanoma cells upon endoplasmic reticulum stress through autophagy. Autophagy. 2015;11(7):975–994. doi: 10.1080/15548627.2015.1049800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Tuoheti Z, et al. RIPK1 polymorphisms alter the susceptibility to cervical Cancer among the Uyghur population in China. BMC Cancer. 2020;20(1):299. doi: 10.1186/s12885-020-06779-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Akimoto M, et al. Antidiabetic adiponectin receptor agonist AdipoRon suppresses tumour growth of pancreatic cancer by inducing RIPK1/ERK-dependent necroptosis. Cell Death Dis. 2018;9(8):804. doi: 10.1038/s41419-018-0851-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Di Grazia A, et al. The fragile X mental retardation protein regulates RIPK1 and colorectal cancer resistance to necroptosis. Cell Mol Gastroenterol Hepatol. 2021;11(2):639–658. doi: 10.1016/j.jcmgh.2020.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nicolè L, et al. Necroptosis-driving genes RIPK1, RIPK3 and MLKL-p are associated with intratumoral CD3(+) and CD8(+) T cell density and predict prognosis in hepatocellular carcinoma. J Immunother Cancer. 2022 doi: 10.1136/jitc-2021-004031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kondylis V, Pasparakis M. RIP Kinases in liver cell death, inflammation and cancer. Trends Mol Med. 2019;25(1):47–63. doi: 10.1016/j.molmed.2018.10.007. [DOI] [PubMed] [Google Scholar]
  • 22.Ting AT, Bertrand MJM. More to Life than NF-kappaB in TNFR1 Signaling. Trends Immunol. 2016;37(8):535–545. doi: 10.1016/j.it.2016.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cao L, Mu W. Necrostatin-1 and necroptosis inhibition: pathophysiology and therapeutic implications. Pharmacol Res. 2021;163:105297. doi: 10.1016/j.phrs.2020.105297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Feyen D, et al. Increasing short-term cardiomyocyte progenitor cell (CMPC) survival by necrostatin-1 did not further preserve cardiac function. Cardiovasc Res. 2013;99(1):83–91. doi: 10.1093/cvr/cvt078. [DOI] [PubMed] [Google Scholar]
  • 25.Liang S, Nian Z, Shi K. Inhibition of RIPK1/RIPK3 ameliorates osteoclastogenesis through regulating NLRP3-dependent NF-kappaB and MAPKs signaling pathways. Biochem Biophys Res Commun. 2020;526(4):1028–1035. doi: 10.1016/j.bbrc.2020.03.177. [DOI] [PubMed] [Google Scholar]
  • 26.Annibaldi A, et al. Ubiquitin-mediated regulation of RIPK1 kinase activity independent of IKK and MK2. Mol Cell. 2018;69(4):566–580.e5. doi: 10.1016/j.molcel.2018.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Darding M, Meier P. IAPs: guardians of RIPK1. Cell Death Differ. 2012;19(1):58–66. doi: 10.1038/cdd.2011.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Varfolomeev E, Vucic D. RIP1 post-translational modifications. Biochem J. 2022;479(9):929–951. doi: 10.1042/BCJ20210725. [DOI] [PubMed] [Google Scholar]
  • 29.Katsuya K, et al. High-throughput screening for linear ubiquitin chain assembly complex (LUBAC) selective inhibitors using homogenous time-resolved fluorescence (HTRF)-based assay system. SLAS Discov. 2018;23(10):1018–1029. doi: 10.1177/2472555218793066. [DOI] [PubMed] [Google Scholar]
  • 30.Tanzer MC, et al. Combination of IAP antagonist and IFNγ activates novel caspase-10- and RIPK1-dependent cell death pathways. Cell Death Differ. 2017;24(3):481–491. doi: 10.1038/cdd.2016.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhang X, Dowling JP, Zhang J. RIPK1 can mediate apoptosis in addition to necroptosis during embryonic development. Cell Death Dis. 2019;10(3):245. doi: 10.1038/s41419-019-1490-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhang T, et al. Necroptosis pathways in tumorigenesis. Semin Cancer Biol. 2022 doi: 10.1016/j.semcancer.2022.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yin Z, et al. RIPK1 is a negative mediator in Aquaporin 1-driven triple-negative breast carcinoma progression and metastasis. NPJ Breast Cancer. 2021;7(1):53. doi: 10.1038/s41523-021-00261-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yu YQ, et al. SMYD2 targets RIPK1 and restricts TNF-induced apoptosis and necroptosis to support colon tumor growth. Cell Death Dis. 2022;13(1):52. doi: 10.1038/s41419-021-04483-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fang T, et al. Alterations in necroptosis during ALDH2mediated protection against high glucoseinduced H9c2 cardiac cell injury. Mol Med Rep. 2018;18(3):2807–2815. doi: 10.3892/mmr.2018.9269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Liang W, et al. A novel damage mechanism: contribution of the interaction between necroptosis and ROS to high glucose-induced injury and inflammation in H9c2 cardiac cells. Int J Mol Med. 2017;40(1):201–208. doi: 10.3892/ijmm.2017.3006. [DOI] [PubMed] [Google Scholar]
  • 37.Yang SH, et al. Nec-1 alleviates cognitive impairment with reduction of Abeta and tau abnormalities in APP/PS1 mice. EMBO Mol Med. 2017;9(1):61–77. doi: 10.15252/emmm.201606566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Delanghe T, Dondelinger Y, Bertrand MJM. RIPK1 Kinase-dependent death: a symphony of phosphorylation events. Trends Cell Biol. 2020;30(3):189–200. doi: 10.1016/j.tcb.2019.12.009. [DOI] [PubMed] [Google Scholar]
  • 39.Katsuya K, et al. Small-molecule inhibitors of linear ubiquitin chain assembly complex (LUBAC), HOIPINs, suppress NF-kappaB signaling. Biochem Biophys Res Commun. 2019;509(3):700–706. doi: 10.1016/j.bbrc.2018.12.164. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

12672_2023_725_MOESM1_ESM.pdf (1MB, pdf)

Additional file 1: Fig S1. RIPK1 is down-regulated in tumor cells of lymphoma patients. A Heatmap of 3 groups of DEGs (CLL, DLBCL, and FL vs. normal, respectively). Chronic lymphocytic leukemia (CCL, N=5), follicular lymphoma (FL, N=10), and diffuse large B-cell patients (DLBCL, N=8) vs. normal donors (N=6). The cutoff was set at p-adjust < 0.001 and log2 fold change > 3. B Volcano plot of DEGs of FL vs. normal. Follicular lymphoma (FL, N=10) vs. normal donors (N=6). A cutoff of p-adjust < 0.001 and log2 fold change > 3. Fig S2. RIPK1 inhibitor promotes the proliferation of lymphoma cells A CCK8 assay determined cell proliferation of L428 lines. Cells were seeded onto 96-well plates and pre-treated with Nec (30 μM) for 24 h, and then, cell growth status was detected by CCK8 assay every 12 h. N=3, *P<0.05, **P<0.01, ***P<0.001 by two-way ANOVA. B Cell viability of L428 cell lines. Cells in A were seeded onto 96-well plates and pre-treated with Nec (30 μM) for 24 h, and then, cell growth status was detected every 12 h. N=3, *P<0.05, ***P<0.001 by two-way ANOVA. C PI staining determined the cell cycle distribution of L428 cell lines and the statistical results. ~5000 cells were pre-treated with Nec (30 μM) for 24 h and then collected for PI staining. D PI staining determined the cell cycle distribution of GM12878/L428 cell lines and the statistical results. ~5000 cells were pre-treated with RIPK1-IN-7 (IN-7, 5 nM) for 24 h and then collected for PI staining. N=6, ***P<0.001 by student’s t-test. E Annexin V-FITC/PI double-labeled FACS determined the cell death of L428 cell lines. Cells in A were used for experiments. Fig S3. HOIPIN-1 inhibits the proliferation of lymphoma cells by blocking RIPK1 function. A CCK8 assay determined cell proliferation of U2932 lines. Cells were seeded onto 96-well plates and pre-treated with HOIPIN-1 (HOI, 10 μM) for 24 h, and then, cell growth status was detected by CCK8 assay every 12 h. N=3, *P<0.05, **P<0.01, ***P<0.001 by two-way ANOVA. B Cell viability of U2932 cell lines. Cells in A were seeded onto 96-well plates and pre-treated with HOI (10 μM) for 24 h, and then, cell growth status was detected every 12 h. N=3, *P<0.05, ***P<0.001 by two-way ANOVA. CD. PI staining determined the cell cycle distribution of U2932 cell lines and the statistical results D. ~5000 cells were pre-treated with HOI at the indicated dose for 24 h and then collected for PI staining. N=6, *P<0.05, ***P<0.001 by one-way ANOVA.E. Annexin V-FITC/PI double-labeled FACS determined the cell death of TMD8 cell lines. Cells in Fig. 3B were used for experiments.


Articles from Discover. Oncology are provided here courtesy of Springer

RESOURCES