ORIGINAL RESEARCH article

Front. Immunol.

Sec. Cancer Immunity and Immunotherapy

Volume 16 - 2025 | doi: 10.3389/fimmu.2025.1591615

This article is part of the Research TopicMechanisms Regulating Immune Evasion by B-cell LymphomaView all articles

Response gene to complement 32 promotes tumorigenesis by mediating DNA damage repair and inhibits CD8+ T cells infiltration in diffuse large B-cell lymphoma

Provisionally accepted
Xiyuan  ZhangXiyuan Zhang1,2Tiange  LuTiange Lu2Chunlei  ShiChunlei Shi3Mengfei  DingMengfei Ding2Ling  WangLing Wang1Xinting  HuXinting Hu2Xin  WangXin Wang2,4*
  • 1Department of Hematology, University of Health and Rehabilitation Sciences, Qingdao Central Hospital, Qingdao, Shandong Province, China
  • 2Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, China
  • 3Department of Hematology, Qingdao Traditional Chinese Medicine Hospital, Qingdao Hiser Hospital, Qingdao University, Qingdao, Shandong Province, China
  • 4Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China

The final, formatted version of the article will be published soon.

Background: Response gene to complement 32 (RGC32), a complement activation-inducible factor broadly expressed in normal human tissues, has been implicated in tumorigenesis through its dysregulated expression in various malignancies and its involvement in critical oncogenic processes. Despite its established roles in cancer biology, RGC32 remains uncharacterized in diffuse large B-cell lymphoma (DLBCL). This study provides the first comprehensive investigation of RGC32 expression patterns and functional contributions to DLBCL pathogenesis, elucidating its potential as a novel therapeutic target or prognostic biomarker in this disease. Methods: Immunohistochemical (IHC) staining of RGC32 was performed on specimens from 32 Reactive hyperplasia lymphoid (RHL) patients and 80 DLBCL patients. To evaluate the role of RGC32 in DLBCL, lentivirus vectors either encoding shRGC32 or shControl were transfected into DLBCL cell lines. RNA-sequencing (RNA-seq) analysis was performed between shRGC32 and shControl stably transfected OCI-LY1 cells and functional enrichment analyses used gene ontology (GO) and kyoto encylopaedia of genes and genomes (KEGG). In order to explored its functions in vivo, xenograft models were established by subcutaneously injecting shRGC32 and shControl transfected DLBCL cells into SCID beige mice. Results: Immunohistochemical analysis revealed RGC32 overexpression in DLBCL tissues contrast with RHL, and was associated advanced Ann Arbor stage (p = 0.043), B symptoms (p = 0.020), and poor progression-free survival (p = 0.015) and overall survival (p = 0.035). Functional studies demonstrated that RGC32 knockdown via shRNA significantly suppressed DLBCL cell proliferation in vitro and in vivo, with xenograft models showing reduced tumor growth and Ki-67 expression. RNA-seq analysis linked RGC32 depletion to downregulation of cell proliferation and impaired DNA damage repair (DDR) mechanisms. Western blot showed RGC32 knockdown could suppress ATM/ATR/CHK1 pathway and increase the tumor mutational burden (TMB). Furthermore, after inhibition of RGC32, infiltration of CD8+ T cells was increased in DLBCL tumor microenvironment (TME). Conclusions: This study highlights that RGC32 is a novel molecule in DLBCL progression and might be a potential therapeutic target for DLBCL therapy.

Keywords: Response gene to complement 32 (RGC32), Diffuse large B cell lymphoma (DLBCL), Cell Cycle, DNA damage repair (DDR), CD8+ T cells

Received: 11 Mar 2025; Accepted: 09 Jun 2025.

Copyright: © 2025 Zhang, Lu, Shi, Ding, Wang, Hu and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

* Correspondence: Xin Wang, Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, 250100, Shandong Province, China

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