1
|
Wang B, Zhang Q, Wang L, Su H, Zhou L, Zhang R, Wan Q. ADAMDEC1 promotes cervical squamous cell carcinoma by enhancing the JAK/STAT signaling pathway through modulation of TYMP. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6009-6020. [PMID: 39625490 DOI: 10.1007/s00210-024-03680-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/25/2024] [Indexed: 04/11/2025]
Abstract
Cervical squamous cell carcinoma (CSCC) poses a significant health challenge, especially in developing countries. Identifying novel prognostic biomarkers and potential drug targets is crucial for improving CSCC management. We analyzed ADAMDEC1 expression patterns in CSCC using various bioinformatic datasets. Clinical samples from CSCC patients were evaluated for ADAMDEC1 and TYMP levels through Western blot and qRT-PCR. Multiple in vitro techniques, including flow cytometry, CCK-8, Transwell, Western blot, wound healing assays, and Co-IP, were employed to investigate the role and molecular pathways associated with ADAMDEC1. Our findings reveal that ADAMDEC1 is significantly overexpressed in CSCC tissues compared to adjacent healthy tissues. Functional assays demonstrated that ADAMDEC1 overexpression significantly enhances cell proliferation, migration, and invasion in vitro, while concurrently inhibiting apoptosis. Conversely, the knockdown of ADAMDEC1 reversed these effects, leading to reduced cell proliferation and increased apoptosis. Mechanistically, we identified a direct interaction between ADAMDEC1 and TYMP, which activates the JAK/STAT signaling pathway in CSCC cells. This activation was confirmed through Western blot analysis, indicating increased phosphorylation of JAK1 and STAT3 in response to ADAMDEC1 overexpression. ADAMDEC1 promotes CSCC progression by modulating the JAK/STAT pathway through regulation of TYMP. These findings suggest that ADAMDEC1 could serve as a molecular biomarker for early diagnosis and targeted therapy in CSCC.
Collapse
Affiliation(s)
- Beibei Wang
- Department of Gynecological Oncology, The 1st Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Qingsong Zhang
- Department of Gynecological Oncology, The 1st Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Lihua Wang
- Department of Gynecological Oncology, The 1st Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Huiwen Su
- Department of Gynecological Oncology, The 1st Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Li Zhou
- Department of Gynecological Oncology, The 1st Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Rong Zhang
- The 1st Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Qiangkun Wan
- Department of Radiotherapy, The 1st Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China.
| |
Collapse
|
2
|
Lugassy J, Abdala-Saleh N, Jarrous G, Turky A, Saidemberg D, Ridner-Bahar G, Berger N, Bar-On D, Taura T, Wilson D, Karin N. Development of DPP-4-resistant CXCL9-Fc and CXCL10-Fc chemokines for effective cancer immunotherapy. Proc Natl Acad Sci U S A 2025; 122:e2501791122. [PMID: 40238455 PMCID: PMC12037015 DOI: 10.1073/pnas.2501791122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/10/2025] [Indexed: 04/18/2025] Open
Abstract
CXCR3 is a chemokine receptor for three ligands: CXCL9, CXCL10, and CXCL11. Accumulating evidence, including data presented here, suggests that the interaction between CXCL9/CXCL10 and CXCR3 not only attracts CXCR3+ T cells but also promotes the induction of IFNγ-high effector/cytotoxic CD4+ and CD8+ T cells, establishing a CXCL9/10-CXCR3-IFNγ self-amplifying cycle that promotes efficient cancer cell killing. One of the homeostatic mechanisms that may limit this cycle is the cleavage of the two N-terminal amino acids of these chemokines by Dipeptidyl Peptidase IV (DPP-4). The modified chemokines retain their ability to bind CXCR3 but no longer activate it, becoming competitive antagonists to native CXCL9/CXCL10. To develop a DPP-4-resistant variant, we combined biochemical analysis with computational modeling, demonstrating that the addition of N-terminal glutamine (Q) to CXCL9-Fc and CXCL10-Fc rendered them fully active CXCR3 agonists, yet resistant to DPP-4 cleavage. Preclinical evaluations imply that they offer significant therapeutic potential in cancer immunotherapy.
Collapse
Affiliation(s)
- Jennie Lugassy
- Department of Immunology, Faculty of Medicine, Technion, Haifa3525422, Israel
| | - Noor Abdala-Saleh
- Department of Immunology, Faculty of Medicine, Technion, Haifa3525422, Israel
| | - Ghada Jarrous
- Department of Immunology, Faculty of Medicine, Technion, Haifa3525422, Israel
| | - Abeer Turky
- Department of Immunology, Faculty of Medicine, Technion, Haifa3525422, Israel
| | - Daniel Saidemberg
- Research and Development, Teva Pharmaceutical Industries, Ltd., Netanya4250419, Israel
| | - Gabriela Ridner-Bahar
- Research and Development, Teva Pharmaceutical Industries, Ltd., Netanya4250419, Israel
| | - Nir Berger
- Research and Development, Teva Pharmaceutical Industries, Ltd., Netanya4250419, Israel
| | - Dana Bar-On
- Research and Development, Teva Pharmaceutical Industries, Ltd., Netanya4250419, Israel
| | - Tetsuya Taura
- Biologics Discovery, Teva Pharmaceutical Industries Ltd., Redwood City, CA94063
| | - David Wilson
- Biologics Discovery, Teva Pharmaceutical Industries Ltd., Redwood City, CA94063
| | - Nathan Karin
- Department of Immunology, Faculty of Medicine, Technion, Haifa3525422, Israel
| |
Collapse
|
3
|
Zafar M, Sweis N, Kapoor H, Gantt G. Advances and Challenges in the Treatment of HPV-Associated Lower Genital Tract Cancers by Immune Checkpoint Blockers: Insights from Basic and Clinical Science. Cancers (Basel) 2025; 17:1260. [PMID: 40282436 PMCID: PMC12026392 DOI: 10.3390/cancers17081260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Human papillomavirus (HPV)-related lower genital cancers, including cervical cancer, anal squamous cell carcinoma (SCC), vaginal cancer, vulvar cancer, and penile cancer, pose a significant health burden, with approximately 45,000 new cases diagnosed annually. Current effective treatment modalities include chemoradiotherapy, systemic chemotherapy, and immune checkpoint inhibitors (ICIs). The tumor microenvironment in HPV-related cancers is characterized by immune evasion mechanisms, including the modulation of immune checkpoints such as PD-L1/PD-1. HPV oncoproteins E5, E6, and E7 play crucial roles in this process, altering the expression of immune inhibitory molecules and the recruitment of immune cells. ICIs, such as programmed cell death protein 1 (PD-1) inhibitors, have shown efficacy in enhancing the immune response against HPV-associated tumors by blocking proteins that allow cancer cells to evade immune surveillance. Recent studies have demonstrated that HPV-positive tumors exhibit a more favorable response to ICI-based therapies compared to HPV-negative tumors. The integration of ICIs into treatment regimens for HPV-related cancers has been supported by several clinical trials. The inclusion of ICIs in the treatment approach for HPV-related lower genital cancers presents a promising opportunity for improving patient outcomes. Ongoing research and clinical trials are advancing our understanding of the immune microenvironment and the therapeutic potential of immunotherapy for these cancers.
Collapse
|
4
|
Li J, Ma Y, Wu Q, Ping P, Li J, Xu X. The potential role of HPV oncoproteins in the PD-L1/PD-1 pathway in cervical cancer: new perspectives on cervical cancer immunotherapy. Front Oncol 2024; 14:1488730. [PMID: 39735605 PMCID: PMC11671370 DOI: 10.3389/fonc.2024.1488730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Cervical cancer (CC) is a common malignant tumour of the female reproductive system that is highly harmful to women's health. The efficacy of traditional surgery, radiotherapy and chemotherapy is limited, especially for recurrent and metastatic CC. With continuous progress in diagnostic and treatment technology, immunotherapy has become a new approach for treating CC and has become a new therapy for recurrent and metastatic CC. However, immunotherapy is not effective for all patients with CC. Therefore, factors related to immunotherapy efficacy in CC patients have become the focus of researchers. High-risk human papillomavirus (HPV) infection is an important factor that drives CC development and affects its progression and prognosis. Increasing attention has been given to the mechanism of the E5, E6 and E7 proteins, which are encoded by the HPV gene, in the occurrence and development of CC and their interaction with programmed cell death ligand-1/programmed cell death-1 (PD-L1/PD-1). Although some preliminary studies have been conducted on these topics, a comprehensive and systematic review of these topics is not available. This review comprehensively summarizes related articles from journals with impact factors greater than 3 and published in the past 5 years; it also reviews studies on the mechanism of HPV and CC, the mechanism of PD-L1/PD-1 axis regulation in CC, and the mechanism by which the interaction between HPV-related oncoproteins and the PD-L1/PD-1 pathway affects the development and prognosis of CC. This study provides theoretical support for the use of immunotherapies for CC, provides a basis for the selection of specific medications that target different HPV-related proteins, and provides a new perspective for the discovery of new immunotherapy targets for CC.
Collapse
Affiliation(s)
| | | | | | | | - Juan Li
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Dalian Medical
University, Dalian, China
| | - Xiaoying Xu
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Dalian Medical
University, Dalian, China
| |
Collapse
|
5
|
Yang J, Long X, Li S, Zhou M, Hu LN. The correlation between vaginal pathogens and high-risk human papilloma virus infection: a meta-analysis of case-control studies. Front Oncol 2024; 14:1423118. [PMID: 39640283 PMCID: PMC11618108 DOI: 10.3389/fonc.2024.1423118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/21/2024] [Indexed: 12/07/2024] Open
Abstract
Background Systematic study on the relationship between vaginal microbiota and high-risk human papillomavirus infection (HR-HPV) is limited. Hence, the aim of this study is to investigate the correlation between vaginal microbiota and HR-HPV infection through a meta-analysis of case-control studies. Methods Chinese Journal Full-text database, Wanfang database, PubMed database, VIP Chinese Science and Technology Journal database, Web of Science, ScienceDirect, ProQuest, JSTOR, Wiley, and IEEE Xplore were synthetically searched for studies about the correlation between vaginal microbiota and HR-HPV infection. Revman 5.3 software was used to assess the relationship between vaginal microbiota and HPV infection through meta-analysis. Finally, forest map was used to calculate the results and funnel plot was applied to test the publication bias. Results Fourteen independent studies were admitted in this study, containing a total of 21, 446 women in gynecological outpatients. Compared with HR-HPV negative group, the prevalence of bacterial vaginosis (BV) [odds ratio (OR)=2.45, 95% confidence intervals (CI): 1.83-3.27, P<0.00001], Ureaplasma urealyticum (UU) (OR=1.38, 95% CI: 1.23-1.54, P<0.00001), and Chlamydia trachomatis (CT) (OR=3.53, 95% CI: 2.82-4.41, P<0.00001) increased in HR-HPV positive group through meta-analysis, while, there was no significant difference in the prevalence of trichomonal vaginitis (TV) (OR=1.69, 95% CI: 0.97-2.96, P=0.06) and vulvovaginal candidiasis (VVC) (OR=0.91, 95% CI: 0.54-1.51, P=0.71. Conclusions Vaginal pathogens are closely related to HR-HPV infection. When BV, UU, and CT are abnormal, the risk of HR-HPV infection is increased.
Collapse
Affiliation(s)
- Jun Yang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
| | - Xin Long
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
| | - Sijing Li
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
| | - Min Zhou
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing, China
| | - Li-Na Hu
- The Center for Reproductive Medicine, Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Joint International Research Lab for Reproduction and Development, Ministry of Education, Chongqing, China
- Reproduction and Stem Cell Therapy Research Center of Chongqing, Chongqing, China
| |
Collapse
|
6
|
Xin X, Li Z, Yan X, Liu T, Li Z, Chen Z, Yan X, Zeng F, Hou L, Zhang J. Hepatocyte-specific Smad4 deficiency inhibits hepatocarcinogenesis by promoting CXCL10/CXCR3-dependent CD8 +- T cell-mediated anti-tumor immunity. Theranostics 2024; 14:5853-5868. [PMID: 39346534 PMCID: PMC11426237 DOI: 10.7150/thno.97276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/18/2024] [Indexed: 10/01/2024] Open
Abstract
Rationale: Sma mothers against decapentaplegic homologue 4 (Smad4) is a key mediator of the transforming growth factor β (TGF-β) pathway and plays complex and contradictory roles in hepatocellular carcinoma (HCC). However, the specific role of Smad4 in hepatocytes in regulating hepatocarcinogenesis remains poorly elucidated. Methods: A diethylnitrosamine/carbon tetrachloride-induced HCC model was established in mice with hepatocyte-specific Smad4 deletion (AlbSmad4-/-) and liver tumorigenesis was monitored. Immune cell infiltration was examined by immunofluorescence and fluorescence activated cell sorting (FACS). Cytokine secretion, glycolysis, signal pathway, and single-cell RNA sequencing were analysed for mechanism. Results: AlbSmad4-/- mice exhibited significantly fewer and smaller liver tumor nodules, less fibrosis, reduced myeloid-derived suppressor cell infiltration and increased CD8+ T cell infiltration. Smad4 deletion in hepatocytes enhanced C-X-C motif ligand 10 (CXCL10) secretion, promoting tumor necrosis factor-α (TNF-α) production in CD8+ T cells. The loss of Smad4 activated the CXCL10/mammalian target of rapamycin (mTOR)/lactate dehydrogenase A (LDHA) pathway, which increased glycolytic activity in CD8+ T cells. HCC patients with high Smad4 expression exhibited decreased CD8+ T cell infiltration and altered glycolysis. Conclusion: Our results demonstrate that Smad4 in hepatocytes promotes hepatocarcinogenesis and is a potential and candidate target for the prevention and therapy of HCC.
Collapse
Affiliation(s)
- Xin Xin
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Xuanxuan Yan
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Ting Liu
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong province, China
| | - Zuyin Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Zhuomiaoyu Chen
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, China
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan province, China
| | - Lingling Hou
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Jinhua Zhang
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| |
Collapse
|
7
|
Liu Y, Niu M, Luo Y, Pan M, Hong S. DNA damage response and inflammatory response: Two traffic lights for HPVs on the road to transformation. J Med Virol 2024; 96:e29815. [PMID: 39073137 DOI: 10.1002/jmv.29815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/17/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
Human papillomaviruses (HPVs) are non-enveloped double-stranded DNA viruses. When HPV infection persists, infected tissues can develop many HPV-related diseases such as cervical cancer and head and neck squamous cell carcinoma. To establish their persistent infection, HPVs have evolved mechanisms to manipulate the host cellular processes such as DNA damage response (DDR), which includes homologous recombination, nonhomologous end joining, and microhomology-mediated end joining. Additionally, HPVs utilize host inflammatory processes to facilitate their life cycles. Here, we bridge the concepts of DDR and inflammatory response, and discuss how HPV proteins orchestrate a sophisticated manipulation of DDR and inflammation to promote their viral replication, ultimately fostering the progression of infected cells towards oncogenic transformation to malignancy.
Collapse
Affiliation(s)
- Yanfei Liu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Mengda Niu
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Ying Luo
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Min Pan
- Department of Otorhinolaryngology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shiyuan Hong
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Wang W, Wan F, Yu T, Wu S, Cui X, Xiang C, Li M, Liu Q, Lin C. Microvesicles-delivering Smad7 have advantages over microvesicles in suppressing fibroblast differentiation in a model of Peyronie's disease. BMC Biotechnol 2024; 24:40. [PMID: 38849776 PMCID: PMC11162046 DOI: 10.1186/s12896-024-00866-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND This study compared the differences of microvesicles (MVs) and microvesicles-delivering Smad7 (Smad7-MVs) on macrophage M1 polarization and fibroblast differentiation in a model of Peyronie's disease (PD). METHODS Overexpression of Smad7 in rat BMSCs was obtained by pCMV5-Smad7 transfection. MVs were collected from rat BMSCs using ultracentrifugation. In cells, 100 µg/mL of MVs or Smad7-MVs were used to treat the 100 ng/mL of lipopolysaccharide (LPS)-induced RAW264.7 cells or 10 ng/mL of recombinant transforming growth factor-β1 (TGF-β1)-induced fibroblasts. The pro-inflammatory cytokines and markers of M1 macrophages were measured in RAW264.7 cells, and the migration and markers of fibroblast differentiation were measured in fibroblasts. In rats, 50 µg of MVs or Smad7-MVs were used to treat the TGF-β1-induced animals. The pathology of tunica albuginea (TA), the markers of M1 macrophages and fibroblast differentiation in the TA were measured. RESULTS The MVs or Smad7-MVs treatment suppressed the LPS-induced macrophage M1 polarization and TGF-β1-induced fibroblast differentiation. Moreover, the Smad7-MVs treatment decreased the fibroblast differentiation compared with the MVs treatment. In the TGF-β1-induced TA of rats, MVs or Smad7-MVs treatment ameliorated the TA fibrosis by suppressing the macrophage M1 polarization and fibroblast differentiation. There was no significance on the M1-polarized macrophages between the MVs treatment and the Smad7-MVs treatment. Meanwhile, the Smad7-MVs treatment had an edge in terms of suppressing the fibroblast differentiation in the TGF-β1-induced PD model compared with the MVs treatment. CONCLUSIONS This study demonstrated that Smad7-MVs treatment had advantages over MVs treatment in suppressing of fibroblast differentiation in a model of PD.
Collapse
Affiliation(s)
- Wenting Wang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Fengchun Wan
- Organ Transplant Center, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Tianxi Yu
- School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Shuang Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Xin Cui
- School of Clinical Medicine, Weifang Medical University, Weifang, 261042, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Chongjun Xiang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
- The 2nd Medical College of Binzhou Medical University, Yantai, 264003, China
| | - Monong Li
- Department of Urology, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Qingzuo Liu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| | - Chunhua Lin
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| |
Collapse
|
9
|
Lu MM, Yang Y. Exosomal PD-L1 in cancer and other fields: recent advances and perspectives. Front Immunol 2024; 15:1395332. [PMID: 38726017 PMCID: PMC11079227 DOI: 10.3389/fimmu.2024.1395332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
PD-1/PD-L1 signaling is a key factor of local immunosuppression in the tumor microenvironment. Immune checkpoint inhibitors targeting PD-1/PD-L1 signaling have achieved tremendous success in clinic. However, several types of cancer are particularly refractory to the anti-PD-1/PD-L1 treatment. Recently, a series of studies reported that IFN-γ can stimulate cancer cells to release exosomal PD-L1 (exoPD-L1), which possesses the ability to suppress anticancer immune responses and is associated with anti-PD-1 response. In this review, we introduce the PD-1/PD-L1 signaling, including the so-called 'reverse signaling'. Furthermore, we summarize the immune treatments of cancers and pay more attention to immune checkpoint inhibitors targeting PD-1/PD-L1 signaling. Additionally, we review the action mechanisms and regulation of exoPD-L1. We also introduce the function of exoPD-L1 as biomarkers. Finally, we review the methods for analyzing and quantifying exoPD-L1, the therapeutic strategies targeting exoPD-L1 to enhance immunotherapy and the roles of exoPD-L1 beyond cancer. This comprehensive review delves into recent advances of exoPD-L1 and all these findings suggest that exoPD-L1 plays an important role in both cancer and other fields.
Collapse
Affiliation(s)
- Man-Man Lu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Zhao W, Li Q, Wen S, Li Y, Bai Y, Tian Z. Novel biomarkers of inflammation-associated immunity in cervical cancer. Front Oncol 2024; 14:1351736. [PMID: 38532933 PMCID: PMC10964772 DOI: 10.3389/fonc.2024.1351736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/12/2024] [Indexed: 03/28/2024] Open
Abstract
Background Cervical cancer (CC) is a highly malignant gynecological cancer with a direct causal link to inflammation, primarily resulting from persistent high-risk human papillomavirus (HPV) infection. Given the challenges in early detection and mid to late-stage treatment, our research aims to identify inflammation-associated immune biomarkers in CC. Methods Using a bioinformatics approach combined with experimental validation, we integrated two CC datasets (GSE39001 and GSE63514) in the Gene Expression Omnibus (GEO) to eliminate batch effects. Immune-related inflammation differentially expressed genes (DGEs) were obtained by R language identification. Results This analysis identified 37 inflammation-related DEGs. Subsequently, we discussed the different levels of immune infiltration between CC cases and controls. Weighted gene co-expression network analysis (WGCNA) identified seven immune infiltration-related modules in CC. We identified 15 immune DEGs associated with inflammation at the intersection of these findings. In addition, we constructed a protein interaction network using the String database and screened five hub genes using "CytoHubba": CXC chemokine ligand 8 (CXCL8), CXC chemokine ligand 10 (CXCL10), CX3C chemokine receptor 1 (CX3CR1), Fc gamma receptors 3B (FCGR3B), and SELL. The expression of these five genes in CC was determined by PCR experiments. In addition, we assessed their diagnostic value and further analyzed the association of immune cells with them. Conclusions Five inflammation- and immune-related genes were identified, aiming to provide new directions for early diagnosis and mid to late-stage treatment of CC from multiple perspectives.
Collapse
Affiliation(s)
- Weihong Zhao
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qi Li
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Songquan Wen
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yaqin Li
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Ying Bai
- Department of Obstetrics and Gynecology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhiyu Tian
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
11
|
Li J, Zhang Y, Luo B. The programed death-1/programed death ligand-1 axis and its potential as a therapeutic target for virus-associated tumours. Rev Med Virol 2024; 34:e2486. [PMID: 37905387 DOI: 10.1002/rmv.2486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/07/2023] [Accepted: 10/22/2023] [Indexed: 11/02/2023]
Abstract
As an important and serious condition impacting human health, the diagnosis, and treatment of tumours is clinically vital because tumour cell immune escape sustains tumour development. Programed death ligand-1 (PD-L1) on tumour cell surfaces binds to the programed death-1 (PD-1), inhibits T cell activation, and induces apoptosis, and incapacitates cells. This allows tumour cells to evade recognition and clearance by the immune system, thereby permitting tumour occurrence, and development and poor prognosis outcomes in patients with tumours. Currently, anti-PD-1/PD-L1 immunotherapy has become pivotal in tumour treatment. Pathogens, especially viruses, are important factors which induce many tumours. In this article, we examine associations between Epstein-Barr virus, human papilloma virus, hepatitis B virus, hepatitis C virus, and human immunodeficiency virus type 1-related tumours and PD-1/PD-L1 axis.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Bing Luo
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Fernandez-Avila L, Castro-Amaya AM, Molina-Pineda A, Hernández-Gutiérrez R, Jave-Suarez LF, Aguilar-Lemarroy A. The Value of CXCL1, CXCL2, CXCL3, and CXCL8 as Potential Prognosis Markers in Cervical Cancer: Evidence of E6/E7 from HPV16 and 18 in Chemokines Regulation. Biomedicines 2023; 11:2655. [PMID: 37893029 PMCID: PMC10604789 DOI: 10.3390/biomedicines11102655] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Cervical cancer (CC) is a serious global health issue, and it is well-known that HPV infection is the main etiological factor that triggers carcinogenesis. In cancer, chemokine ligands and receptors are involved in tumor cell growth, metastasis, leukocyte infiltration, and angiogenesis; however, information on the role played by E6/E7 of HPV16/18 in the modulation of chemokines is very limited. Therefore, this study aimed to determine whether chemokines are differentially expressed in CC-derived cell lines; if E6/E7 oncoproteins from HPV16 and 18 are capable of mediating chemokine expression, what is the expression profile of chemokines in tissues derived from CC and what is their impact on the overall survival of patients with this pathology? For this purpose, RNA sequencing and real-time PCR were performed on SiHa, HeLa, and C33A tumorigenic cell lines, on the non-tumorigenic HaCaT cells, and the E6/E7 HPV-transduced HaCaT cell models. Furthermore, chemokine expression and survival analysis were executed on 304 CC and 22 normal tissue samples from The Cancer Genome Atlas (TCGA) repository. The results demonstrate that CXCL1, CXCL2, CXCL3, and CXCL8 are regulated by E6/E7 of HPV16 and 18, are overexpressed in CC biopsies, and that their higher expression is related to a worse prognostic survival.
Collapse
Affiliation(s)
- Leonardo Fernandez-Avila
- Programa de Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara 44340, Jalisco, Mexico;
| | - Aribert Maryosly Castro-Amaya
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara 44340, Jalisco, Mexico;
| | - Andrea Molina-Pineda
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C., Guadalajara 44270, Jalisco, Mexico; (A.M.-P.); (R.H.-G.)
- Consejo Nacional de Ciencia y Tecnología, CONAHCYT, Mexico City 03940, Mexico
| | - Rodolfo Hernández-Gutiérrez
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C., Guadalajara 44270, Jalisco, Mexico; (A.M.-P.); (R.H.-G.)
| | - Luis Felipe Jave-Suarez
- Programa de Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara 44340, Jalisco, Mexico;
| | - Adriana Aguilar-Lemarroy
- Programa de Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara 44340, Jalisco, Mexico;
| |
Collapse
|
13
|
Olivera C, Mosmann JP, Anna AN, Bettucci Ferrero GN, Paira DA, Ferreyra FN, Martinez MS, Motrich RD, Cuffini CG, Saka HA, Rivero VE. Expression of HPV-16 E6 and E7 oncoproteins alters Chlamydia trachomatis developmental cycle and induces increased levels of immune regulatory molecules. Front Cell Infect Microbiol 2023; 13:1214017. [PMID: 37743859 PMCID: PMC10516566 DOI: 10.3389/fcimb.2023.1214017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Infection with Human Papillomavirus (HPV) is a recognized risk factor for Chlamydia trachomatis (CT) infection and vice versa. Coinfection of HPV and CT in women is a very common and usually asymptomatic finding that has been linked to increased risk of cervical cancer. It has been demonstrated that CT facilitates the entry of multiple high risk HPV genotypes, leading to damage of the mucosal barrier and interfering with immune responses and viral clearance, which ultimately favours viral persistence and malignant transformation. Although the facilitating effects elicited by CT infection on viral persistence have been reported, little is known about the consequences of HPV infection on CT development. Methods Herein, we took advantage of a genetically modified human cervical cell line co-expressing HPV-16 major oncogenic proteins E6 and E7, as an experimental model allowing to investigate the possible effects that HPV infection would have on CT development. Results and discussion Our results show that CT infection of HPV-16 E6E7 expressing cells induced an upregulation of the expression of E6E7 oncoproteins and host cell inhibitory molecules PD-L1, HVEM and CD160. Additionally, smaller chlamydial inclusions and reduced infectious progeny generation was observed in E6E7 cells. Ultrastructural analysis showed that expression of E6 and E7 did not alter total bacterial counts within inclusions but resulted in increased numbers of reticulate bodies (RB) and decreased production of infectious elementary bodies (EB). Our results indicate that during CT and HPV coinfection, E6 and E7 oncoproteins impair RB to EB transition and infectious progeny generation. On the other hand, higher expression of immune inhibitory molecules and HPV-16 E6E7 are cooperatively enhanced in CT-infected cells, which would favour both oncogenesis and immunosuppression. Our findings pose important implications for clinical management of patients with HPV and CT coinfection, suggesting that screening for the mutual infection could represent an opportunity to intervene and prevent severe reproductive health outcomes, such as cervical cancer and infertility.
Collapse
Affiliation(s)
- Carolina Olivera
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Jessica P. Mosmann
- Instituto de Virología “Dr. José M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Ailen N. Anna
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gloria N. Bettucci Ferrero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Daniela A. Paira
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Fernando N. Ferreyra
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María S. Martinez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Rubén D. Motrich
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cecilia G. Cuffini
- Instituto de Virología “Dr. José M. Vanella”, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Héctor Alex Saka
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Virginia E. Rivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
14
|
Hu R, Jahan MS, Tang L. ExoPD-L1: an assistant for tumor progression and potential diagnostic marker. Front Oncol 2023; 13:1194180. [PMID: 37736550 PMCID: PMC10509558 DOI: 10.3389/fonc.2023.1194180] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 08/11/2023] [Indexed: 09/23/2023] Open
Abstract
The proliferation and function of immune cells are often inhibited by the binding of programmed cell-death ligand 1 (PD-L1) to programmed cell-death 1 (PD-1). So far, many studies have shown that this combination poses significant difficulties for cancer treatment. Fortunately, PD-L1/PD-1 blocking therapy has achieved satisfactory results. Exosomes are tiny extracellular vesicle particles with a diameter of 40~160 nm, formed by cells through endocytosis. The exosomes are a natural shelter for many molecules and an important medium for information transmission. The contents of exosomes are composed of DNA, RNA, proteins and lipids etc. They are crucial to antigen presentation, tumor invasion, cell differentiation and migration. In addition to being present on the surface of tumor cells or in soluble form, PD-L1 is carried into the extracellular environment by tumor derived exosomes (TEX). At this time, the exosomes serve as a medium for communication between tumor cells and other cells or tissues and organs. In this review, we will cover the immunosuppressive role of exosomal PD-L1 (ExoPD-L1), ExoPD-L1 regulatory factors and emerging approaches for quantifying and detecting ExoPD-L1. More importantly, we will discuss how targeted ExoPD-L1 and combination therapy can be used to treat cancer more effectively.
Collapse
Affiliation(s)
- Rong Hu
- School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Md Shoykot Jahan
- School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Lijun Tang
- School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
15
|
Qualliotine JR, Nakagawa T, Rosenthal SB, Sadat S, Ballesteros-Merino C, Xu G, Mark A, Nasamran A, Gutkind JS, Fisch KM, Guo T, Fox BA, Khan Z, Molinolo AA, Califano JA. A Network Landscape of HPVOPC Reveals Methylation Alterations as Significant Drivers of Gene Expression via an Immune-Mediated GPCR Signal. Cancers (Basel) 2023; 15:4379. [PMID: 37686653 PMCID: PMC10486378 DOI: 10.3390/cancers15174379] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/17/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
HPV-associated oropharynx carcinoma (HPVOPC) tumors have a relatively low mutational burden. Elucidating the relative contributions of other tumor alterations, such as DNA methylation alterations, alternative splicing events (ASE), and copy number variation (CNV), could provide a deeper understanding of carcinogenesis drivers in this disease. We applied network propagation analysis to multiple classes of tumor alterations in a discovery cohort of 46 primary HPVOPC tumors and 25 cancer-unaffected controls and validated our findings with TCGA data. We identified significant overlap between differential gene expression networks and all alteration classes, and this association was highest for methylation and lowest for CNV. Significant overlap was seen for gene clusters of G protein-coupled receptor (GPCR) pathways. HPV16-human protein interaction analysis identified an enriched cluster defined by an immune-mediated GPCR signal, including CXCR3 cytokines CXCL9, CXCL10, and CXCL11. CXCR3 was found to be expressed in primary HPVOPC, and scRNA-seq analysis demonstrated CXCR3 ligands to be highly expressed in M2 macrophages. In vivo models demonstrated decreased tumor growth with antagonism of the CXCR3 receptor in immunodeficient but not immunocompetent mice, suggesting that the CXCR3 axis can drive tumor proliferation in an autocrine fashion, but the effect is tempered by an intact immune system. In conclusion, methylation, ASE, and SNV alterations are highly associated with network gene expression changes in HPVOPC, suggesting that ASE and methylation alterations have an important role in driving the oncogenic phenotype. Network analysis identifies GPCR networks, specifically the CXCR3 chemokine axis, as modulators of tumor-immune interactions that may have proliferative effects on primary tumors as well as a role for immunosurveillance; however, CXCR3 inhibition should be used with caution, as these agents may both inhibit and stimulate tumor growth considering the competing effects of this cytokine axis. Further investigation is needed to explore opportunities for targeted therapy in this setting.
Collapse
Affiliation(s)
- Jesse R. Qualliotine
- Department of Otolaryngology—Head and Neck Surgery, University of California San Diego, La Jolla, CA 92093, USA
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Takuya Nakagawa
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Medicine, Chiba University, Chiba 263-8522, Japan
| | - Sara Brin Rosenthal
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Sayed Sadat
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Guorong Xu
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Adam Mark
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Art Nasamran
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - J. Silvio Gutkind
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Kathleen M. Fisch
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Theresa Guo
- Department of Otolaryngology—Head and Neck Surgery, University of California San Diego, La Jolla, CA 92093, USA
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Bernard A. Fox
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR 97213, USA
| | - Zubair Khan
- Department of Otolaryngology—Head and Neck Surgery, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alfredo A. Molinolo
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Joseph A. Califano
- Department of Otolaryngology—Head and Neck Surgery, University of California San Diego, La Jolla, CA 92093, USA
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
16
|
Zhang C, Gu L, Xiao J, Jin F. Knockdown of RBM15 inhibits tumor progression and the JAK-STAT signaling pathway in cervical cancer. BMC Cancer 2023; 23:684. [PMID: 37474926 PMCID: PMC10360283 DOI: 10.1186/s12885-023-11163-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/08/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND RNA binding motif protein 15 (RBM15), a writer of N6-methyladenosine (m6A) methylation, contributes significantly to the development of various tumors. However, the function of RBM15 in cervical cancer (CC) has not been determined. METHODS Based on the GSE9750, GSE63514, and m6A datasets, m6A-related differentially expressed genes (DEGs) were screened out. The hub genes were identified by generating a Protein-Protein Interaction (PPI) network. RT-qPCR was conducted to assess the mRNA expression of hub genes. CCK8, scratch wound healing, and transwell assays were utilized to examine the influence of RBM15 on HeLa and SiHa cells. Tumor xenograft models were used to assess the effects of RBM15 on tumorigenesis. A mechanistic analysis of RBM15 in CC tumors was conducted using the GeneCards and Coxpresdb databases, followed by a Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and the pathway-related genes were subsequently validated using Western blotting. RESULTS Five DEGs were screened, including WTAP, RBM15, CBLL1, and YTHDC2. Among them, WTAP, RBM15, CBLL1, and YTHDC2 were hub genes and can be used as biomarkers for CC. RBM15 expression was considerably increased, while WTAP, CBLL1, and YTHDC2 were significantly downregulated. Knockdown of RBM15 significantly suppressed the proliferation, invasion, and migration of CC cells and tumorigenesis. Moreover, knockdown of RBM15 significantly reduced the expression levels of proteins related to the JAK-STAT pathway. CONCLUSIONS Knockdown of RBM15 inhibited the progression of CC cells, which probably by inhibiting the JAK-STAT pathway pathway.
Collapse
Affiliation(s)
- Chunnian Zhang
- Department of Gynecology, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Ganzhou City, 341000, Jiangxi Province, China.
| | - Liqin Gu
- Department of Gynecology, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Ganzhou City, 341000, Jiangxi Province, China
| | - Juan Xiao
- Department of Gynecology, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Ganzhou City, 341000, Jiangxi Province, China
| | - Feng Jin
- Department of Gynecology, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Ganzhou City, 341000, Jiangxi Province, China
| |
Collapse
|
17
|
Gombos G, Németh N, Pös O, Styk J, Buglyó G, Szemes T, Danihel L, Nagy B, Balogh I, Soltész B. New Possible Ways to Use Exosomes in Diagnostics and Therapy via JAK/STAT Pathways. Pharmaceutics 2023; 15:1904. [PMID: 37514090 PMCID: PMC10386711 DOI: 10.3390/pharmaceutics15071904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Exosomes have the potential to be the future of personalized diagnostics and therapy. They are nano-sized particles between 30 and 100 nm flowing in the extracellular milieu, where they mediate cell-cell communication and participate in immune system regulation. Tumor-derived exosomes (TDEs) secreted from different types of cancer cells are the key regulators of the tumor microenvironment. With their immune suppressive cargo, TDEs prevent the antitumor immune response, leading to reduced effectiveness of cancer treatment by promoting a pro-tumorigenic microenvironment. Involved signaling pathways take part in the regulation of tumor proliferation, differentiation, apoptosis, and angiogenesis. Signal transducers and activators of transcription factors (STATs) and Janus kinase (JAK) signaling pathways are crucial in malignancies and autoimmune diseases alike, and their potential to be manipulated is currently the focus of interest. In this review, we aim to discuss exosomes, TDEs, and the JAK/STAT pathways, along with mediators like interleukins, tripartite motif proteins, and interferons.
Collapse
Affiliation(s)
- Gréta Gombos
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary
| | - Nikolett Németh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary
| | - Ondrej Pös
- Comenius University Science Park, 841 04 Bratislava, Slovakia
- Geneton Ltd., 841 04 Bratislava, Slovakia
| | - Jakub Styk
- Comenius University Science Park, 841 04 Bratislava, Slovakia
- Geneton Ltd., 841 04 Bratislava, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Gergely Buglyó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary
| | - Tomas Szemes
- Comenius University Science Park, 841 04 Bratislava, Slovakia
- Geneton Ltd., 841 04 Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 841 01 Bratislava, Slovakia
| | - Ludovit Danihel
- 3rd Surgical Clinic, Faculty of Medicine, Comenius University and Merciful Brothers University Hospital, 811 08 Bratislava, Slovakia
| | - Bálint Nagy
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary
- Comenius University Science Park, 841 04 Bratislava, Slovakia
| | - István Balogh
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Beáta Soltész
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary
| |
Collapse
|
18
|
Deng Y, Song Y, Du Q, Wang CC, Li H, Sui Y, Zhang Y, Tang T. Anti-HPV16 oncoproteins siRNA therapy for cervical cancer using a novel transdermal peptide PKU12. Front Oncol 2023; 13:1175958. [PMID: 37350944 PMCID: PMC10282752 DOI: 10.3389/fonc.2023.1175958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/15/2023] [Indexed: 06/24/2023] Open
Abstract
In this study, an innovative transdermal peptide, #PKU12, was developed based on transdermal peptide TD-1, and the anti-tumor effect of PKU12-based siRNA against HPV was investigated in vivo. Furthermore, transcriptome differences between PKU12 + siRNA treatment and control groups were compared to assess treatment effects. The top five upregulated and downregulated genes identified by RNA sequencing were further subjected to survival analysis. The present study, for the first time, showed that this novel peptide could enhance the transdermal delivery of the siRNA targeting HPV16 L1, E6, and E7. PKU12-based siRNA delivery significantly repressed the mRNA expression levels of HPV16 L1, E6, and E7 in the SiHa xenograft tumors and attenuated tumor growth as well. The RNA-sequencing results showed that a total of 586 DEGs were detected in the PKU12 + siRNA-treated tumor tissues compared to the control tumor tissues. The GSEA analysis revealed that DEGs were inversely associated with the HIF-1 signaling pathway, the TNF signaling pathway, the AGE-RAGE signaling pathway, the NF-kappa B signaling pathway, ferroptosis, the IL-17 signaling pathway, ovarian steroidogenesis, and rheumatoid arthritis. Further functional enrichment analysis revealed that DEGs were significantly enriched in several key pathways, including cytokine-cytokine receptor interaction, the TNF signaling pathway, and the IL-17 signaling pathway. High expression of MYH1, MYH4, FGG, DEPP1, and ZBTB16 was associated with shorter overall survival of patients with cervical cancer; high expression of SULT1E1, RAB3C, CXCR3, and PROX2 was associated with longer overall survival of patients with cervical cancer. In conclusion, the transdermal peptide PKU12 is potentially a good candidate for a siRNA delivery vehicle for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Yan Deng
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yi Song
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Quan Du
- The State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chi Chiu Wang
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Science, and School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hu Li
- Department of Gynecology, Pan Yu Central Hospital, Guangzhou, China
- Cancer Institute, Panyu Central Hospital, Guangzhou, China
| | - Yi Sui
- Department of Nutrition, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuying Zhang
- Department of Obstetrics and Gynaecology, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Tao Tang
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Gynecology, Pan Yu Central Hospital, Guangzhou, China
- Department of Obstetrics and Gynaecology, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| |
Collapse
|
19
|
Duan Y, Zhang X, Ying H, Xu J, Yang H, Sun K, He L, Li M, Ji Y, Liang T, Bai X. Targeting MFAP5 in cancer-associated fibroblasts sensitizes pancreatic cancer to PD-L1-based immunochemotherapy via remodeling the matrix. Oncogene 2023:10.1038/s41388-023-02711-9. [PMID: 37156839 DOI: 10.1038/s41388-023-02711-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023]
Abstract
Highly desmoplastic and immunosuppressive tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC) contributes to tumor progression and resistance to current therapies. Clues targeting the notorious stromal environment have offered hope for improving therapeutic response whereas the underlying mechanism remains unclear. Here, we find that prognostic microfibril associated protein 5 (MFAP5) is involved in activation of cancer-associated fibroblasts (CAFs). Inhibition of MFAP5highCAFs shows synergistic effect with gemcitabine-based chemotherapy and PD-L1-based immunotherapy. Mechanistically, MFAP5 deficiency in CAFs downregulates HAS2 and CXCL10 via MFAP5/RCN2/ERK/STAT1 axis, leading to angiogenesis, hyaluronic acid (HA) and collagens deposition reduction, cytotoxic T cells infiltration, and tumor cells apoptosis. Additionally, in vivo blockade of CXCL10 with AMG487 could partially reverse the pro-tumor effect from MFAP5 overexpression in CAFs and synergize with anti-PD-L1 antibody to enhance the immunotherapeutic effect. Therefore, targeting MFAP5highCAFs might be a potential adjuvant therapy to enhance the immunochemotherapy effect in PDAC via remodeling the desmoplastic and immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Yi Duan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Xiaozhen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Honggang Ying
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Jian Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Hanshen Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Kang Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Lihong He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Muchun Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Yongtao Ji
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China
- Cancer Center, Zhejiang University, Hangzhou, 310000, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China.
- Cancer Center, Zhejiang University, Hangzhou, 310000, China.
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Provincial Innovation Center for The Study of Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for The Study of Hepatobiliary & Pancreatic Diseases, Zhejiang University, Hangzhou, 310000, China.
- Cancer Center, Zhejiang University, Hangzhou, 310000, China.
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
20
|
Rizwi FA, Abubakar M, Puppala ER, Goyal A, Bhadrawamy CV, Naidu VGM, Roshan S, Tazneem B, Almalki WH, Subramaniyan V, Rawat S, Gupta G. Janus Kinase-Signal Transducer and Activator of Transcription Inhibitors for the Treatment and Management of Cancer. J Environ Pathol Toxicol Oncol 2023; 42:15-29. [PMID: 37522565 DOI: 10.1615/jenvironpatholtoxicoloncol.2023045403] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
According to the World Health Organization (WHO), cancer is the second-highest cause of mortality worldwide, killing nearly 9.6 million people annually. Despite the advances in diagnosis and treatment during the last couple of decades, it remains a serious concern due to the limitations of currently available cancer management strategies. Therefore, alternative strategies are highly required to overcome these glitches. In addition, many etiological factors such as environmental and genetic factors initiate the activation of the Janus kinase (JAK)-signal transducer and activator of the transcription (STAT) pathway. This aberrant activation of the JAK-STAT pathway has been reported in various disease states, including inflammatory conditions, hematologic malignancies, and cancer. For instance, many patients with myeloproliferative neoplasms carry the acquired gain-of-function JAK2 V617F somatic mutation. This knowledge has dramatically improved our understanding of pathogenesis and has facilitated the development of therapeutics capable of suppressing the constitutive activation of the JAK-STAT pathway. Our aim is not to be expansive but to highlight emerging ideas towards preventive therapy in a modern view of JAK-STAT inhibitors. A series of agents with different specificities against different members of the JAK family of proteins is currently undergoing evaluation in clinical trials. Here we give a summary of how JAK-STAT inhibitors function and a detailed review of current clinical drugs for managing cancer as a new therapeutic approach.
Collapse
Affiliation(s)
- Fahim Anwar Rizwi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - Md Abubakar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - Eswara Rao Puppala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Ch Veera Bhadrawamy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - V G M Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Sila Katamur, Halugurisuk P.O-Changsari, Kamrup, Assam, India-781101
| | - S Roshan
- Deccan School of Pharmacy, Hyderabad, India
| | - B Tazneem
- Deccan School of Pharmacy, Hyderabad, India
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, MONASH University, Malaysia
| | - Sushama Rawat
- Nirma University, Institute of Pharmacy, Ahmedabad, Gujarat 382481, India; School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura 302017, Jaipur, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura 302017, Jaipur, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
21
|
Pu D, Liu D, Li C, Chen C, Che Y, Lv J, Yang Y, Wang X. A novel ten-gene prognostic signature for cervical cancer based on CD79B-related immunomodulators. Front Genet 2022; 13:933798. [PMID: 36406115 PMCID: PMC9666757 DOI: 10.3389/fgene.2022.933798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/09/2022] [Indexed: 01/25/2023] Open
Abstract
The identification of immune-related prognostic biomarkers opens up the possibility of developing new immunotherapy strategies against tumors. In this study, we investigated immune-related biomarkers in the tumor microenvironment to predict the prognosis of cervical cancer (CC) patients. ESTIMATE and CIBERSORT algorithms were used to calculate the abundance of tumor-infiltrating immune cells (TICs) and the amount of immune and stromal components in cervical samples (n = 309) from The Cancer Genome Atlas. Ten immune-related differentially expressed genes associated with CC survival were identified via intersection analyses of multivariate Cox regression and protein-protein interactions. CD79B was chosen for further study, and its prognostic value and role in anti-CC immune functions were analyzed. Differential expression analysis and qRT-PCR validation both revealed that CD79B expression was down-regulated in CC tissues. Survival analysis suggested that a high level of CD79B expression was associated with good prognosis. In the clinical correlation analysis, CD79B expression was found to be related to primary therapy outcome, race, histological type, degree of cell differentiation, disease-specific survival, and progression-free interval. GSEA showed that the function and pathway of CD79B were mainly related to immune activities. Meanwhile, CD79B expression was correlated with 10 types of TICs. Based on CD79B-associated immunomodulators, a novel immune prognostic signature consisting of 10 genes (CD96, LAG3, PDCD1, TIGIT, CD27, KLRK1, LTA, PVR, TNFRSF13C, and TNFRSF17) was established and validated as possessing good independent prognostic value for CC patients. Finally, a nomogram to predict personalized 3- and 5-year overall survival probabilities in CC patients was built and validated. In summary, our findings demonstrated that CD79B might be a potential prognostic biomarker for CC. The 10-gene prognostic signature independently predicted the overall survival of patients with CC, which could improve individualized treatment and aid clinical decision-making.
Collapse
Affiliation(s)
- Dan Pu
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Dan Liu
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China,Department of Gynecology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Can Li
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Chunyan Chen
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yuxin Che
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jiaoyan Lv
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yang Yang
- Department of Medical Basic Experimental Teaching Center, China Medical University, Shenyang, China,*Correspondence: Yang Yang, ; Xuelian Wang,
| | - Xuelian Wang
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China,*Correspondence: Yang Yang, ; Xuelian Wang,
| |
Collapse
|
22
|
Zhang J, Dong D, Wei Q, Ren L. CXCL10 serves as a potential serum biomarker complementing SCC-Ag for diagnosing cervical squamous cell carcinoma. BMC Cancer 2022; 22:1052. [PMID: 36207693 PMCID: PMC9547472 DOI: 10.1186/s12885-022-10142-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 09/27/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cervical squamous cell carcinoma (CESC) is the most common histological type of cervical cancer which is the major cause of death in women worldwide. Although squamous cell carcinoma antigen (SCC-Ag) is widely used to detect CESC, it is not sensitive and specific enough to predict the disease. METHODS We investigated serum CXC motif chemokine 10 (CXCL10) as potential diagnostic biomarker in detecting CESC in this study. Serum levels of CXCL10 and SCC-Ag were measured by ELISA or automated immunoassay in 345 participants, including 189 patients with different stages of CESC, 75 patients with cervical intraepithelial neoplasia, and 81 healthy individuals. Performances of CXCL10 and SCC-Ag as single biomarkers were analyzed by the ROC curves. The changes of serum levels of CXCL10 and SCC-Ag in 10 longitudinal followed-up CESC patients with partial response (PR) during chemoradiotherapy or chemotherapy were evaluated. RESULTS The two markers showed similar diagnostic capacity in distinguishing both CESC early stage from healthy controls (AUCCXCL10 = 0.740, AUCSCC-Ag = 0.710) and all CESC from healthy controls (AUCCXCL10 = 0.775, AUCSCC-Ag =0.793). Moreover, CXCL10 showed ability in distinguishing cervical intraepithelial neoplasia from healthy control (AUCCXCL10 = 0.727) and cervical cancer SCC-Ag-negative from healthy control. (AUCCXCL10 = 0.739). The combination of CXCL10 and SCC-Ag displayed significant improvement of AUCs than individual SCC-Ag or CXCL10 in the analysis groups (healthy vs all cervical cancer, healthy vs cervical cancer early stage). The AUCs were improved to 0.877 (AUCSCC-Ag = 0.793, P < 0.05) to distinguish healthy controls from all CESC and 0.828(AUCSCC-Ag = 0.710, P < 0.05) to distinguish healthy controls from CESC early stage by the combination of the two markers, respectively. Significant differences of serum CXCL10 levels were found between CESC patients at late tumor stage and CESC patients at early tumor stage (P < 0.01). Serum CXCL10 levels of the CESC patients who had partial response after treatment significantly decreased during treatment (P = 0.013), whose consistent and inconsistent frequency with the response were the same as serum SCC-Ag levels. CONCLUSIONS The results indicated that CXCL10 is a potential serum biomarker complementing SCC-Ag in prediction of CESC. CXCL10 showed ability in the diagnosis of SCC-Ag negative CESC and the combination of CXCL10 and SCC-Ag inhibited improved performance compared with SCC-Ag alone.
Collapse
Affiliation(s)
- Jingya Zhang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin, 300060, P.R. China
| | - Dong Dong
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin, 300060, P.R. China
| | - Qian Wei
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin, 300060, P.R. China
| | - Li Ren
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin, 300060, P.R. China.
| |
Collapse
|
23
|
Tantengco OAG, Richardson LS, Radnaa E, Kammala AK, Kim S, Medina PMB, Han A, Menon R. Modeling ascending Ureaplasma parvum infection through the female reproductive tract using vagina-cervix-decidua-organ-on-a-chip and feto-maternal interface-organ-on-a-chip. FASEB J 2022; 36:e22551. [PMID: 36106554 PMCID: PMC9500016 DOI: 10.1096/fj.202200872r] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/08/2022] [Accepted: 09/02/2022] [Indexed: 09/02/2023]
Abstract
Genital mycoplasmas can break the cervical barrier and cause intraamniotic infection and preterm birth. This study developed a six-chamber vagina-cervix-decidua-organ-on-a-chip (VCD-OOC) that recapitulates the female reproductive tract during pregnancy with culture chambers populated by vaginal epithelial cells, cervical epithelial and stromal cells, and decidual cells. Cells cultured in VCD-OOC were characterized by morphology and immunostaining for cell-specific markers. We transferred the media from the decidual cell chamber of the VCD-OOC to decidual cell chamber in feto-maternal interface organ-on-a-chip (FMi-OOC), which contains the fetal membrane layers. An ascending Ureaplasma parvum infection was created in VCD-OOC. U. parvum was monitored for 48 h post-infection with their cytotoxicity (LDH assay) and inflammatory effects (multiplex cytokine assay) in the cells tested. An ascending U. parvum infection model of PTB was developed using CD-1 mice. The cell morphology and expression of cell-specific markers in the VCD-OOC mimicked those seen in lower genital tract tissues. U. parvum reached the cervical epithelial cells and decidua within 48 h and did not cause cell death in VCD-OOC or FMi-OOC cells. U. parvum infection promoted minimal inflammation, while the combination of U. parvum and LPS promoted massive inflammation in the VCD-OOC and FMi-OOC cells. In the animal model, U. parvum vaginal inoculation of low-dose U. parvum did not result in PTB, and even a high dose had only some effects on PTB (20%). However, intra-amniotic injection of U. parvum resulted in 67% PTB. We report the colonization of U. parvum in various cell types; however, inconsistent, and low-grade inflammation across multiple cell types suggests poor immunogenicity induced by U. parvum.
Collapse
Affiliation(s)
- Ourlad Alzeus G. Tantengco
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
- Biological Models Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Lauren S. Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Enkhtuya Radnaa
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Sungjin Kim
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Paul Mark B. Medina
- Biological Models Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
24
|
Single-Cell Transcriptome Analysis Reveals Different Immune Signatures in HPV- and HPV + Driven Human Head and Neck Squamous Cell Carcinoma. J Immunol Res 2022; 2022:2079389. [PMID: 36157879 PMCID: PMC9507777 DOI: 10.1155/2022/2079389] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/16/2022] [Indexed: 12/01/2022] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is a significant health problem and related to poor long-term outcomes, indicating more research to be done to deeply understand the underlying pathways. Objective This current study aimed in the assessment of the viral- (especially human papilloma virus [HPV]) and carcinogen-driven head and neck squamous cell carcinoma (HNSCC) microenvironment based on single-cell sequencing analysis. Methods Data were downloaded from GEO database (GSE139324), including 131224 cells from 18 HP- HNSCC patients and 8 HPV+ HNSCC patients. Following data normalization, all highly variable genes in single cells were identified, and batch correction was applied. Differentially expressed genes were identified using Wilcoxon rank sum test. A gene enrichment analysis was performed in each cell cluster using KEGG analysis. Single-cell pseudotime trajectories were constructed with MONOCLE (version 2.6.4). Cell-cell interactions were analyzed with CellChat R package. Additionally, cell-cell communication patterns in key signal pathways were compared in different tissue groups. A hidden Markov model (HMM) was used to predict gene expression states (on or off) throughout pseudotime. Five-year overall survival outcomes were compared in both HPV+ and HPV- subsets. Results 20,978 high-quality individual cells passed quality control. RNA-seq data were used from 522 HNSCC primary tumor samples. 1,137 differentially expressed genes between HPV+ and HPV- HNSCC patients were investigated. 96 differentially expressed genes were associated with overall survival and highly enriched in B cell associated biological process. Cell composition differed between types of samples. MHC-I, MHC-II, and MIF signaling pathways were found to be most relevant. Within these pathways, some cells were either signal receiver or signal sender, depending on sample type, respectively. Six genes were obtained, AREG and TGFBI (upregulation), CD27, CXCR3, MS4A1, and CD19 (downregulation), whose expression and HPV types were highly associated with worse overall survival. AREG and TGFBI were pDC marker genes, CXCR3 and CD27 were significantly expressed in T cell-related cells, while MS4A1 and CD19 were mainly expressed in B naïve cells. Conclusions This study revealed dynamic changes in cell percentage and heterogeneity of cell subtypes of HNSCC. AREG, TGFBI, CD27, CXCR3, MS4A1, and CD19 were associated with worse overall survival in HPV-related HNSCC. Especially B-cell related pathways were revealed as particularly relevant in HPV-related HNSCC. These findings are a basis for the development of biomarkers and therapeutic targets in respective patients.
Collapse
|
25
|
Xu J, Huang Z, Wang Y, Xiang Z, Xiong B. Identification of Novel Tumor Microenvironment Regulating Factor That Facilitates Tumor Immune Infiltration in Cervical Cancer. Front Oncol 2022; 12:846786. [PMID: 35847936 PMCID: PMC9277773 DOI: 10.3389/fonc.2022.846786] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 06/02/2022] [Indexed: 12/14/2022] Open
Abstract
Cervical cancer is one of the most common gynecologic malignancies and one of the leading causes of cancer-related deaths in women worldwide. There are more than 30 categories of human papillomavirus infections in the genital tract. The recently discovered immune checkpoint suppression is a potential approach to improve clinical outcomes in these patients by altering immune cell function. However, many questions remain unanswered in terms of this method. For example, the proportion of responders is limited and the exact mechanism of action is uncertain. The tumor microenvironment (TME) has long been regarded as having nonnegligible influence on effectiveness of immunotherapy. The programmed cell death protein 1 (PD-1) pathway has received much attention due to its involvement in activating T-cell immune checkpoint responses. Since tumor cells may evade immune detection and become highly resistant to conventional treatments, anti-PD-1/PD-L1 antibodies are preferred as a kind of cancer treatment and many have just been licensed. To provide a theoretical basis for the development of new therapies, investigating the effect of tumor microenvironment on the prognosis of cervical cancer is necessary. In this work, immunological scores obtained from the ESTIMATE algorithm were used to differentiate between patients with high and low immune cell infiltration. We identified 11 immunologically significant differentially expressed genes (DEGs). For example, CXCR3 was found to be an important factor in CD8+ T cell recruitment and tumor immunological infiltration in cervical cancer. These results may lead to novel directions of understanding complex interactions between cancer cells and the tumor microenvironment, as well as new treatment options for cervical cancer.
Collapse
Affiliation(s)
- Jingjing Xu
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Zhe Huang
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Yishu Wang
- Department of Legal English and TOEIC, Adelaide University, North Terrace, SA, Australia
| | - Zhenxian Xiang
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery & Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| |
Collapse
|