1
|
Ingram JL, McQuade VL, Weiss J, Womble JT, Ihrie MD, Zhao K, Francisco D, Theriot B, May K, Kim H, McCravy M, Sauler M, Lugogo NL, Sunday ME, Everitt J, Walker JKL, Tighe RM, Kraft M, Que LG. Leptin augments IL-13-induced airway eotaxins and submucosal eosinophilia in obesity-associated asthma. J Allergy Clin Immunol 2025; 155:819-833.e10. [PMID: 39581293 PMCID: PMC11875949 DOI: 10.1016/j.jaci.2024.10.039] [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: 05/16/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Airway tissue eosinophilia can be an observed feature of obesity-associated type 2 (T2) asthma, but the processes mediating this inflammation are unknown. OBJECTIVE To investigate a process whereby leptin, an adipokine elevated in obesity, potentiates pulmonary eosinophilia and eotaxin production by airway fibroblasts in T2 asthma. METHODS We assessed associations between body mass index and airway eosinophilia as well as leptin and eotaxin production in 82 participants with asthma, 37 of whom exhibited obesity. Cultured human airway fibroblasts and mouse models of chronic allergic airway disease were used to evaluate leptin's effect on eotaxin production and lung eosinophilia. The role of IL-13 receptor alpha 2 (IL-13Rα2) in mediating these processes was examined using specific neutralizing antibodies in vitro. RESULTS In participants with T2 asthma and obesity, we observed that airway tissue eosinophilia did not associate with traditional T2 inflammation metrics such as peripheral and/or bronchoalveolar lavage fluid eosinophil counts or with fractional exhaled nitric oxide. Alternatively, we observed elevated bronchoalveolar lavage fluid leptin and eotaxin-1 levels. In airway fibroblasts from participants with asthma, leptin augmented IL-13-induced eotaxin-1 and eotaxin-3 production and IL13RA2 expression. In mice, elevated leptin promoted airway IL-13Rα2 and eotaxin production by lung fibroblasts and lung tissue eosinophilia following chronic house dust mite allergen exposure. Inhibition of IL-13Rα2 reduced combined leptin and IL-13-stimulated eotaxin secretion by human airway fibroblasts. CONCLUSIONS We identified a potential association explaining airway tissue eosinophil retention in obesity-associated T2 asthma through leptin-mediated enhancement of IL-13-induced eosinophil chemokine production by airway fibroblasts, a process requiring IL-13Rα2.
Collapse
Affiliation(s)
| | | | - Jasmine Weiss
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC
| | - Jack T Womble
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Mark D Ihrie
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Karen Zhao
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Dave Francisco
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Katelynn May
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Haein Kim
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Matthew McCravy
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Maor Sauler
- Department of Internal Medicine, Yale University, New Haven, Conn
| | - Njira L Lugogo
- Department of Medicine, University of Michigan, Ann Arbor, Mich
| | - Mary E Sunday
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Jeffrey Everitt
- Department of Pathology, Duke University Medical Center, Durham, NC
| | | | - Robert M Tighe
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Loretta G Que
- Department of Medicine, Duke University Medical Center, Durham, NC
| |
Collapse
|
2
|
Alobaidi R, Islam N, Olkey T, Haribabu Y, Shamo M, Sykora P, Simbulan-Rosenthal CM, Rosenthal DS. Combinational Inhibition of MEK and AKT Synergistically Induces Melanoma Stem Cell Apoptosis and Blocks NRAS Tumor Growth. Cells 2025; 14:248. [PMID: 39996721 PMCID: PMC11852824 DOI: 10.3390/cells14040248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Malignant melanoma is a lethal skin cancer containing melanoma-initiating cells (MICs), implicated in tumorigenesis, invasion, and drug resistance, and characterized by an elevated expression of stem cell markers, including CD133. siRNA knockdown of CD133 has been previously shown to enhance apoptosis induced by the MEK inhibitor trametinib in melanoma cells. This study investigates the underlying mechanisms of CD133's anti-apoptotic activity in patient-derived BAKP melanoma, harboring the difficult-to-treat NRASQ61K driver mutation, after CRISPR-Cas9 CD133 knockout or Doxycycline (Dox)-inducible re-expression of CD133. CD133 knockout in BAKP cells increased trametinib-induced apoptosis by reducing anti-apoptotic p-AKT and p-BAD and increasing pro-apoptotic BAX. Conversely, Dox-induced CD133 expression diminished apoptosis in trametinib-treated cells, coincident with elevated p-AKT, p-BAD, and decreased activation of BAX and caspase-3. However, trametinib in combination with pan-AKT inhibitor capivasertib reduced cell survival as measured by XTT viability assays and apoptosis and colony formation assays, independent of CD133 status. CD133 may therefore activate a survival pathway wherein (1) increased AKT phosphorylation and activation induces (2) BAD phosphorylation and inactivation, which (3) decreases BAX activation, and (4) reduces caspases-3 activity and caspase-mediated PARP cleavage, leading to apoptosis suppression and drug resistance in melanoma. In vivo mouse xenograft studies using Dox-inducible melanoma cells revealed increased rates of tumor growth after induction of CD133 expression in trametinib-treated +Dox mice, an effect which was synergistically suppressed by combination treatment. Targeting nodes of the AKT and MAPK survival pathways with trametinib and capivasertib highlights the potential for combination therapies for NRAS-mutant melanoma stem cells for the development of more effective treatments for patients with high-risk melanoma.
Collapse
Affiliation(s)
- Ryyan Alobaidi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (R.A.); (N.I.); (T.O.); (Y.H.); (M.S.); (C.M.S.-R.)
- Department of Pathology, King Saud University College of Medicine, Riyadh 11461, Saudi Arabia
| | - Nusrat Islam
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (R.A.); (N.I.); (T.O.); (Y.H.); (M.S.); (C.M.S.-R.)
| | - Toni Olkey
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (R.A.); (N.I.); (T.O.); (Y.H.); (M.S.); (C.M.S.-R.)
| | - Yogameenakshi Haribabu
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (R.A.); (N.I.); (T.O.); (Y.H.); (M.S.); (C.M.S.-R.)
| | - Mathew Shamo
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (R.A.); (N.I.); (T.O.); (Y.H.); (M.S.); (C.M.S.-R.)
| | - Peter Sykora
- Amelia Technologies, LLC, Washington, DC 20001, USA;
| | - Cynthia M. Simbulan-Rosenthal
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (R.A.); (N.I.); (T.O.); (Y.H.); (M.S.); (C.M.S.-R.)
| | - Dean S. Rosenthal
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC 20057, USA; (R.A.); (N.I.); (T.O.); (Y.H.); (M.S.); (C.M.S.-R.)
| |
Collapse
|
3
|
Hakala S, Hämäläinen A, Sandelin S, Giannareas N, Närvä E. Detection of Cancer Stem Cells from Patient Samples. Cells 2025; 14:148. [PMID: 39851576 PMCID: PMC11764358 DOI: 10.3390/cells14020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
The existence of cancer stem cells (CSCs) in various tumors has become increasingly clear in addition to their prominent role in therapy resistance, metastasis, and recurrence. For early diagnosis, disease progression monitoring, and targeting, there is a high demand for clinical-grade methods for quantitative measurement of CSCs from patient samples. Despite years of active research, standard measurement of CSCs has not yet reached clinical settings, especially in the case of solid tumors. This is because detecting this plastic heterogeneous population of cells is not straightforward. This review summarizes various techniques, highlighting their benefits and limitations in detecting CSCs from patient samples. In addition, methods designed to detect CSCs based on secreted and niche-associated signaling factors are reviewed. Spatial and single-cell methods for analyzing patient tumor tissues and noninvasive techniques such as liquid biopsy and in vivo imaging are discussed. Additionally, methods recently established in laboratories, preclinical studies, and clinical assays are covered. Finally, we discuss the characteristics of an ideal method as we look toward the future.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Närvä
- Institute of Biomedicine and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (S.H.); (A.H.); (S.S.); (N.G.)
| |
Collapse
|
4
|
Jing R, Wu N, Zhang Q, Liu J, Zhao Y, Zeng S, Wu S, Wu Y, Yi S. DPP4 promotes an immunoenhancing tumor microenvironment through exhausted CD8+ T cells with activating IL13-IL13RA2 axis in papillary thyroid cancer. Int Immunopharmacol 2025; 145:113760. [PMID: 39662266 DOI: 10.1016/j.intimp.2024.113760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/17/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Papillary thyroid cancer (PTC) is among the most prevalent forms of endocrine malignancy with a rapid rise in incidence rates worldwide; however, the composition and characteristics of its immune microenvironment is poorly understand. Here, this work investigated the precise function of Dipeptidyl peptidase 4 (DPP4) in tumor-infiltrated T cells within PTC by investigating its role in cytokine-mediated signaling pathways. METHODS TCGA and GEO data as well as human PTC specimens confirmed the expression of DPP4 in PTC. The CIBERSORT and TIMER tool were used to analyze the distribution of tumor-infiltrating immune cells in PTC. CD8+ T cells from PTC patient's peripheral blood were cultured and used in a three-dimensional model for direct co-culture with PTC tumors to investigate DPP4 function. RESULTS Bioinformatic analyses has uncovered a significant upregulation of DPP4, which enhances the survival and migration of PTC cells in vitro. DPP4 upregulation significantly correlated with advanced grades, stages, and poor progression-free survival. DPP4 influences immune function and the exhaustion of CD8+ T cells through the IL13-IL13RA2 axis. The inhibition of DPP4 reduces CD8+ T cell exhaustion and IL13 secretion, while also blocking the IL13-IL13RA2 axis, thereby promoting the mesenchymal-to-epithelial transition of PTC cells. CONCLUSION Blocking DPP4 leads to the conversion of exhausted CD8+ T cells with decreased IL13 level, resulting in downregulation of IL13RA2 to promote mesenchymal-to-epithelial transition of PTC cells. This highlights DPP4 as a potential therapeutic target, particularly between CD8+ T cells and PTC cells via IL13-IL13RA2 axis, and represents a novel avenue for combined immunotherapy in PTC.
Collapse
Affiliation(s)
- Ren Jing
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Nan Wu
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Qian Zhang
- Respiratory Medicine, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518118, PR China
| | - Jinlin Liu
- Department of Clinical Laboratory, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Ying Zhao
- Department of Clinical Laboratory, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Shan Zeng
- Department of Pathology, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Shaojie Wu
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China
| | - Yang Wu
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China.
| | - Shijian Yi
- Department of Breast and Thyroid Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen 518116, PR China.
| |
Collapse
|
5
|
Cui B, Yang G, Yan H, Wu S, Wang K, Wang H, Li Y. UBE3C restricts EV-A71 replication by ubiquitination-dependent degradation of 2C. J Virol 2024; 98:e0133524. [PMID: 39212385 PMCID: PMC11494953 DOI: 10.1128/jvi.01335-24] [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: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Ubiquitin modification of viral proteins to degrade or regulate their function is one of the strategies of the host to resist viral infection. Here, we report that ubiquitin protein ligase E3C (UBE3C), an E3 ubiquitin ligase, displayed inhibitory effects on EV-A71 replication. UBE3C knockdown resulted in increased viral protein levels and virus titers, whereas overexpression of UBE3C reduced EV-A71 replication. To explore the mechanism by which UBE3C affected EV-A71 infection, we found that the C-terminal of UBE3C bound to 2C protein and facilitated K33/K48-linked ubiquitination degradation of 2C K268. Moreover, UBE3C lost its ability to degrade 2C K268R and had a diminished inhibitory impact against the replication of recombinant EV-A71-FY-2C K268R. In addition, UBE3C also promoted ubiquitination degradation of the 2C protein of CVB3 and CVA16 and inhibited viral replication. Thus, our findings reveal a novel mechanism that UBE3C acts as an enterovirus host restriction factor, including EV-A71, by targeting the 2C protein. IMPORTANCE The highly conserved 2C protein of EV-A71 is a multifunctional protein and plays a key role in the replication cycle. In this study, we demonstrated for the first time that UBE3C promoted the degradation of 2C K268 via K33/K48-linked ubiquitination, thereby inhibiting viral proliferation. Our findings advance the knowledge related to the roles of 2C in EV-A71 virulence and the ubiquitination pathway in the host restriction of EV-A71 infection.
Collapse
Affiliation(s)
- Boming Cui
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ge Yang
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyan Yan
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuo Wu
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kun Wang
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huiqiang Wang
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuhuan Li
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Li X, Chen M, Cao J, Chen X, Song H, Shi S, He B, Zhang B, Zhang Z. Human umbilical cord mesenchymal stem cell-derived exosomes mitigate diabetic nephropathy via enhancing M2 macrophages polarization. Heliyon 2024; 10:e37002. [PMID: 39286156 PMCID: PMC11402917 DOI: 10.1016/j.heliyon.2024.e37002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Background and objectives Exosomes, which are small nanoscale vesicles capable of secretion, have garnered significant attention in recent years because of their therapeutic potential, particularly in the context of kidney diseases. Notably, human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Exos) are emerging as promising targeted therapies for renal conditions. The aim of this study was to investigate the therapeutic effects of hucMSC-Exos on diabetic kidney disease (DKD) both in vivo and in vitro. Additionally, this study seeks to elucidate cellular and molecular differentials, as well as the expression of relevant signaling pathways, through single-cell RNA sequencing. This endeavor was designed to enhance our understanding of the connection between hucMSC-Exos and the pathogenesis of DKD. Methods and results The study commenced with the extraction and characterization of hucMSC-Exos, including the determination of their concentrations. Animal experiments were conducted to evaluate the therapeutic potential of hucMSC-Exos in a DKD mouse model. Subsequently, single-cell sequencing was employed to investigate the molecular mechanisms underlying the efficacy of extracellular vesicles in ameliorating DKD. These findings were further substantiated by cell-based experiments. Importantly, the results indicate that hucMSC-Exos can impede the progression of DKD in mice, with macrophage activation playing a pivotal role in this process. Conclusions The in vivo experiments conclusively established hucMSC-Exos as a pivotal component in preserving renal function and retarding the progression of DKD. Our utilization of single-cell sequencing technology, in conjunction with in vivo and in vitro experiments, provides compelling evidence that M2 macrophages are instrumental in enhancing the amelioration of diabetic nephropathy.
Collapse
Affiliation(s)
- Xueting Li
- Department of Nephrology, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China
| | - Mingkai Chen
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Jinghe Cao
- Department of Reproductive Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Xinke Chen
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Hui Song
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Shuo Shi
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Baoyu He
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Ziteng Zhang
- Departments of Thoracic Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
- Departments of Thoracic Surgery, Qinghai Red Cross Hospital, Xining, Qinghai, 81000, PR China
| |
Collapse
|
7
|
Wang L, He Z, Li J. Development of a Competitive Chemiluminescent Assay for Quantitative Determination of TP53 Fusion Protein Using Reagent Strips. Appl Biochem Biotechnol 2024; 196:6315-6329. [PMID: 38351430 DOI: 10.1007/s12010-024-04860-3] [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] [Accepted: 02/04/2024] [Indexed: 11/29/2024]
Abstract
Recent studies have shown that almost half of all cancers occur due to DNA damage. For the early diagnosis of cancer, a highly sensitized and swift identification for TP53 is needed since the corresponding TP53 protein is effectively recognized as "the guardian of the genome." To improve the detection sensitivity, numerous analytical methods were previously used for the determination of the TP53 protein, including denaturing high-performance liquid chromatography and enzyme-linked immunosorbent assay (ELISA). Currently, immunochromatographic tests (ICTs) that are simple to use, stable over time, and show low interference are regarded as valuable tools for the quick screening of food and environmental monitoring along with clinical diagnosis. ICTs often have limited sensitivity even if a variety of novel reporters possessing optimum photostability and improved brightness are used as signal-intensity reporters. Compared with N-(4-aminobutyl)-N-(ethylisoluminol) or luminol, a novel luminescent probe, 2',6'-diMethyl-4'-(N-succiniMidyloxycarbonyl) phenyl-10-sulfopropylacridiniuM-9-carboxylate (NSP-DMAE-NHS) has achieved a much higher efficiency, improvement in the biosensor's performance, and amplification of the signal without causing any damage to the biomolecule in terms of its biochemical activity. In this study, the reagent strip method was initially used to detect TP53 fusion protein by combining the advantages of NSP-DMAE-NHS and immunochromatography. In our experiment, the control and study lines on the strips were immobilized through HRP-conjugated goat anti-rabbit IgG and TP53 antigen, respectively. The optimized concentration of the anti-TP53 antibody-NSP-DMAE-NHS immunoconjugates was then added to the TP53 antigen samples. After, the test strips were inserted and left in the aforementioned buffer solution for an additional 20 min. Finally, a lab-made luminous measurement device was used to analyze the corresponding control and study lines on the strips. Under optimized conditions, this method was found to be ultrasensitive, with a wide range of linear responses from 0.0008 ng mL-1 to 1 µg mL-1 and a limit of detection of 0.0008 ng mL-1 (0.013 pM). Thus, a novel competitive chemiluminescent assay based on reagent strips was established for the determination of the TP53 fusion proteins. The strategy has potential applications for ultrasensitive detection in the early diagnosis of cancer.
Collapse
Affiliation(s)
- Linyu Wang
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemical Engineering & Material, Handan University, Handan, 056005, People's Republic of China.
| | - Zhifang He
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemical Engineering & Material, Handan University, Handan, 056005, People's Republic of China
| | - Jianye Li
- Hebei Key Laboratory of Heterocyclic Compounds, College of Chemical Engineering & Material, Handan University, Handan, 056005, People's Republic of China
| |
Collapse
|
8
|
Liu Z, Zheng Y, Yuan M, Zhang G, Yang G. Association of CTACK, IL-2, and IL-13 with increased risk of lung cancer: A Mendelian randomization study. Cytokine 2024; 181:156680. [PMID: 38885591 DOI: 10.1016/j.cyto.2024.156680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND In recent years, relevant studies have reported that inflammatory cytokines are related to the occurrence of cancer. However, the correlation with lung cancer is not clear. This study used the Mendelian random grouping method to investigate the correlation between inflammatory factors and lung cancer in different populations. METHODS We obtained the single nucleotide polymorphisms (SNPs) of inflammatory cytokines through the open database and the SNPs of lung cancer (European and East Asian) through the IEU OpenGWAS project. Inverse variance-weighted (IVW) MR analyses were used to determine the causalities of exposures and outcomes. Supplementary analyses were also performed using weighted median and MR-Egger regressions. Afterward, sensitivity analyses were performed to test the robustness. Search the ChEMBL database for target drugs and indications for CTACK, IL-2, and IL-13. RESULTS By IVW method, we found that CTACK, IL-2, and IL-13 were associated with an increased risk of lung cancer in the European population (CTACK, OR = 1.098, 95 % CI 1.001-1.204, P = 0.047; IL-2, OR = 1.112, 95 % CI 1.009-1.225, P = 0.032; IL-13, OR = 1.068, 95 % CI 1.007-1.132, P = 0.029), while only IL-13 was associated with an increased risk of lung cancer in the East Asian population (IL-13, OR = 1.110, 95 % CI 1.010-1.220, P = 0.030). The weighted median and MR-Egger regression methods were in the same direction as the IVW effect sizes. Furthermore, no evidence of multidirectionality was detected using the MR-Egger intercept as a sensitivity analysis. Currently, there are no approved or phase III studied indications for CTACK, IL-2, and IL-13 targets in lung cancer. CONCLUSION The study outcomes supported that the inflammatory cytokines CTACK, IL-2, and IL-13 increase the risk of lung cancer. There is a lack of indications for drugs in these three targets. We explored the causal relationship between inflammatory cytokines and lung cancer, providing a basis for future cancer prediction models and targets for anti-tumor therapy.
Collapse
Affiliation(s)
- Zishen Liu
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yingying Zheng
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Mengqi Yuan
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ganlin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Guowang Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
LIU ZIYONG, MA TAO, LI JINFANG, REN WEI, ZHANG ZHIXIN. IL13RA2 promotes progression of infantile haemangioma by activating glycolysis and the Wnt/β-catenin signaling pathway. Oncol Res 2024; 32:1453-1465. [PMID: 39220137 PMCID: PMC11361910 DOI: 10.32604/or.2024.048315] [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: 12/04/2023] [Accepted: 02/01/2024] [Indexed: 09/04/2024] Open
Abstract
Background Interleukin 13 receptor subunit alpha 2 (IL13RA2) plays an essential role in the progression of many cancers. However, the role of IL13RA2 in infantile haemangioma (IH) is still unknown. Materials and Methods IL13RA2 expression in IH tissues was analyzed using western blot, qRT-PCR, and immunofluorescence. The role of IL13RA2 in haemangioma-derived endothelial cells (HemECs) was determined following knockdown or overexpression of IL13RA2 using CCK-8, colony formation, apoptosis, wound healing, tubule formation, Transwell, and western blot. Results IL13RA2 expression was upregulated in IH tissues. IL13RA2 overexpression promoted proliferation, migration, and invasion of HemECs and induced glycolysis, which was confirmed with a glycolysis inhibitor. Specifically, IL13RA2 interacted with β-catenin and activated the Wnt/β-catenin pathway in HemECs, which were involved in the above-mentioned effects of IL13RA2. Conclusions These findings revealed that targeting IL13RA2 is a potential therapeutic approach for IH.
Collapse
Affiliation(s)
- ZIYONG LIU
- Department of Cardiothoracic Surgery, 970th Hospital of the People’s Liberation Army, Weihai, 264209, China
| | - TAO MA
- Department of Cardiothoracic Surgery, 970th Hospital of the People’s Liberation Army, Weihai, 264209, China
| | - JINFANG LI
- Department of Cardiac Ultrasound, Weihai Municipal Hospital, Weihai, 264200, China
| | - WEI REN
- Department of Cardiothoracic Surgery, 970th Hospital of the People’s Liberation Army, Weihai, 264209, China
| | - ZHIXIN ZHANG
- Department of Cardiothoracic Surgery, 970th Hospital of the People’s Liberation Army, Weihai, 264209, China
| |
Collapse
|
10
|
Cai X, Gao J, Yan Z, Zhang H, Guo D, Zhang S. MARCH5 promotes hepatocellular carcinoma progression by inducing p53 ubiquitination degradation. J Cancer Res Clin Oncol 2024; 150:303. [PMID: 38861187 PMCID: PMC11166841 DOI: 10.1007/s00432-024-05782-7] [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: 02/09/2024] [Accepted: 05/06/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Human MARCH5 is a mitochondria-localized E3 ubiquitin-protein ligase that is essential for the regulation of mitochondrial dynamics. A large body of evidence suggests that imbalances in mitochondrial dynamics are strongly associated with cancer. However, the expression, biological function and prognostic significance of MARCH5 in hepatocellular carcinoma (HCC) have not been determined. MATERIALS AND METHODS The mRNA and protein expression of MARCH5 in HCC cell lines and tumor tissues was assessed by real-time quantitative PCR, Western blot analysis and immunohistochemistry. The clinical prognostic significance of MARCH5 was evaluated in 135 HCC patients. Knockdown or overexpression of MARCH5 in HCC cells was determined by in vitro cell proliferation, migration and invasion assays, and in vivo tumor growth and metastasis assays. In addition, the intrinsic mechanisms by which MARCH5 regulates HCC cell growth and metastasis were explored. RESULTS MARCH5 was significantly overexpressed in HCC cells and was closely associated with patients' poor postoperative prognosis. In vivo and in vitro experiments revealed that MARCH5 significantly promoted the increase and invasive and migratory ability of hepatocellular carcinoma cells, which was mainly due to the promotion of autophagy by MARCH5. Mechanistic studies revealed that MARCH5 promoted autophagy through ubiquitination degradation of p53 leading to malignant progression of hepatocellular carcinoma. CONCLUSION Our findings suggest that MARCH5 plays a critical oncogenic role in HCC cells, which provides experimental evidence for the use of MARCH5 as a potential target for HCC therapy.
Collapse
Affiliation(s)
- Xin Cai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Liver Transplantation Centre, Zhengzhou, China
- The Main Construction Unit of National Regional Medical Center for Henan Organ Transplantation, Zhengzhou, China
- Henan Research & Development International Joint Laboratory for Organ Transplantation Immunomodulation, Zhengzhou, China
| | - Jie Gao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Liver Transplantation Centre, Zhengzhou, China
- The Main Construction Unit of National Regional Medical Center for Henan Organ Transplantation, Zhengzhou, China
- Henan Research & Development International Joint Laboratory for Organ Transplantation Immunomodulation, Zhengzhou, China
| | - Zhiping Yan
- Zhengzhou Key Laboratory for Hepatobiliary & Pancreatic Diseases and Organ Transplantation, Zhengzhou, China
| | - Huapeng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Liver Transplantation Centre, Zhengzhou, China
- The Main Construction Unit of National Regional Medical Center for Henan Organ Transplantation, Zhengzhou, China
- Henan Research & Development International Joint Laboratory for Organ Transplantation Immunomodulation, Zhengzhou, China
| | - Danfeng Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Liver Transplantation Centre, Zhengzhou, China
- The Main Construction Unit of National Regional Medical Center for Henan Organ Transplantation, Zhengzhou, China
- Henan Research & Development International Joint Laboratory for Organ Transplantation Immunomodulation, Zhengzhou, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Liver Transplantation Centre, Zhengzhou, China.
- The Main Construction Unit of National Regional Medical Center for Henan Organ Transplantation, Zhengzhou, China.
- Henan Research & Development International Joint Laboratory for Organ Transplantation Immunomodulation, Zhengzhou, China.
| |
Collapse
|
11
|
Lei P, Guo Q, Hao J, Liu H, Chen Y, Wu F, He Z, Zhang X, Zhang N, Wen S, Gao W, Wu Y. Exploring the evolving roles and clinical significance of circRNAs in head and neck squamous cell carcinoma. J Cancer 2024; 15:3984-3994. [PMID: 38911371 PMCID: PMC11190751 DOI: 10.7150/jca.96614] [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: 03/25/2024] [Accepted: 05/13/2024] [Indexed: 06/25/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) represents the predominant malignancies in the head and neck region, and has limited therapeutic alternatives. Circular RNAs (circRNAs), a substantial category of non-coding RNA molecules, exert influential roles in human disease development and progression, employing various mechanisms such as microRNA sponging, interaction with RNA-binding proteins, and translational capabilities. Accumulating evidence highlights the differential expression of numerous circRNAs in HNSCC, and numerous dysregulated circRNAs underscore their crucial involvement in malignant advancement and resistance to treatment. This review aims to comprehensively outline the characteristics, biogenesis, and mechanisms of circRNAs, elucidating their functional significance in HNSCC. In addition, we delve into the clinical implications of circRNAs, considering their potential as biomarkers or targets for diagnosis, prognosis, and therapeutic applications in HNSCC. The discussion extends to exploring future challenges in the clinical translation of circRNAs, emphasizing the need for further research.
Collapse
Affiliation(s)
- Pengxiang Lei
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Qingbo Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jiewen Hao
- Department of Otolaryngology Head & Neck Surgery, The Third Hospital of Shanxi Medical University (Shanxi Bethune Hospital), Taiyuan 030032, Shanxi, China
| | - Hui Liu
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Yaofeng Chen
- Department of Otolaryngology Head & Neck Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Feng Wu
- Shenzhen Research Institute, Northwest A&F University, Shenzhen 518000, Guangdong, China
| | - Zhao He
- Shenzhen Research Institute, Northwest A&F University, Shenzhen 518000, Guangdong, China
| | - Xiaolong Zhang
- Shenzhen Research Institute, Northwest A&F University, Shenzhen 518000, Guangdong, China
| | - Nannan Zhang
- Department of Otolaryngology Head & Neck Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Shuxin Wen
- Department of Otolaryngology Head & Neck Surgery, The Third Hospital of Shanxi Medical University (Shanxi Bethune Hospital), Taiyuan 030032, Shanxi, China
| | - Wei Gao
- Department of Otolaryngology Head & Neck Surgery, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China
| | - Yongyan Wu
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
- Department of Otolaryngology Head & Neck Surgery, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China
- Shenzhen Institute of Otolaryngology & Key Laboratory of Otolaryngology, Longgang Otolaryngology Hospital, Shenzhen 518172, Guangdong, China
- Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, Guangdong, China
| |
Collapse
|
12
|
Wang XT, Li L, Zhu Z, Huang YL, Chen HH, Shi ZY, Deng QM, Wu K, Xia LJ, Mai W, Yang JR, Kong FB. SIVA-1 enhances acquired chemotherapeutic drug resistance of gastric cancer in vivo by regulating the ARF/MDM2/p53 pathway. Heliyon 2024; 10:e24394. [PMID: 38312638 PMCID: PMC10834467 DOI: 10.1016/j.heliyon.2024.e24394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
SIVA-1 has been shown to affect apoptotic processes in various different cell lines, and SIVA-1 significantly contributes to the decreased responsiveness of cancer cells to some chemotherapy agents. However, whether SIVA-1 has potential application in gastric cancer remains unknown. Therefore, the objective of this investigation was to clarify the distinct function of SIVA-1 in chemotherapeutic drug resistance within a living murine model with gastric malignancy, and initially elucidate the underlying mechanisms. In an established multidrug-resistant gastric cancer xenograft mouse model, lentivirus, named Lv-SIVA-1, was injected into xenograft tumors, and increased the mRNA and protein expression of endogenous SIVA-1 in tumors. Immunohistochemical assays of xenograft tumor showed that SIVA-1 was significantly upregulated, and the protein expression levels of SIVA-1 were highly increased, as detected by Western blotting. In addition, we detected the role of SIVA-1 in cell proliferation and cell apoptosis in gastric cancer cells by TUNEL and found that SIVA-1 decreased tumor cell apoptosis and promoted tumor growth in vivo. Using a TMT assay between tumor tissues of experimental and control groups, differentially expressed proteins were examined and three potential biomarkers of multidrug resistance (ARF, MDM2, and p53) were screened. We further investigated the molecular mechanism by which SIVA-1 played an efficient role against chemotherapies and found that overexpressed SIVA-1 leads to increased ARF and MDM2 expression and suppressed expression of p53 in tumor tissue. In conclusion, SIVA-1 plays a significant role in the multidrug resistance of gastric tumors. In addition, overexpressed SIVA-1 positively regulates cell proliferation, adjusts cycle progression, and reduces the response to drug treatment for gastric cancer in an ARF/MDM2/p53-dependent manner. This novel research provides a basis for chemical management of gastric cancer through regulation of SIVA-1 expression.
Collapse
Affiliation(s)
- Xiao-Tong Wang
- Departments of Gastrointestinal, Hernia and Enterofistula Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Lei Li
- Departments of Gastrointestinal, Hernia and Enterofistula Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Zhou Zhu
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Yu-Liang Huang
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Huan-Huan Chen
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Zheng-Yi Shi
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Qiao-Ming Deng
- Department of Surgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region, 530023, People’s Republic of China
| | - Kun Wu
- Department of Surgery, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, 530001, People’s Republic of China
| | - Long-Jie Xia
- Department of Cosmetology and Plastic Surgery Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, People’s Republic of China
| | - Wei Mai
- Departments of Gastrointestinal, Hernia and Enterofistula Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
| | - Jian-Rong Yang
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region & Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences & Guangxi Key Laboratory of Eye Health, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
- Jinan University, Guangzhou, Guangdong Province, 510362, People’s Republic of China
| | - Fan-Biao Kong
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Minimally Invasive Technology and Applications Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People’s Republic of China
- Jinan University, Guangzhou, Guangdong Province, 510362, People’s Republic of China
| |
Collapse
|
13
|
Zhao R, Cao G, Zhang B, Wei L, Zhang X, Jin M, He B, Zhang B, He Z, Bie Q. TNF+ regulatory T cells regulate the stemness of gastric cancer cells through the IL13/STAT3 pathway. Front Oncol 2023; 13:1162938. [PMID: 37534250 PMCID: PMC10392945 DOI: 10.3389/fonc.2023.1162938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/16/2023] [Indexed: 08/04/2023] Open
Abstract
Regulatory T cells (Tregs) are an important component of the tumor microenvironment; however, the interaction between Tregs and gastric cancer cells is not completely understood. Recent studies have shown that Tregs participate in cancer cell stemness maintenance. In this study, we performed single-cell RNA sequencing of gastric cancer and adjacent tissues and found that Tregs with high TNF expression were recruited to gastric cancer tissues and were significantly correlated with patient survival. TNF+ Tregs significantly contribute to tumor growth and progression. Our studies have further demonstrated that TNF+ Tregs promote the stemness of gastric cancer cells through the IL13/STAT3 pathway. Therefore, blocking the interaction between TNF+ Tregs and gastric cancer cells may be a new approach in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Rou Zhao
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Guanjie Cao
- Department of Radiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Baogui Zhang
- Colorectal Ward, Department of Gastrointestinal Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Li Wei
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Xiaobei Zhang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Meng Jin
- Hernia and Abdominal Wall Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Baoyu He
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Zhun He
- Department of Thoracic Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Qingli Bie
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| |
Collapse
|