1
|
Lereim RR, Dunn C, Aamdal E, Chauhan SK, Straume O, Guren TK, Kyte JA. Plasma protein dynamics during ipilimumab treatment in metastatic melanoma: associations with tumor response, adverse events and survival. Oncoimmunology 2025; 14:2440967. [PMID: 39703053 DOI: 10.1080/2162402x.2024.2440967] [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: 04/30/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
The immune checkpoint inhibitor ipilimumab provides long term survival in some metastatic melanoma patients, but the majority has no benefit, and may experience serious side effects. Here, we investigated the dynamics of plasma cytokine concentrations and their potential utility for predicting treatment response, adverse events and overall survival (OS) in patients with metastatic melanoma undergoing ipilimumab monotherapy. A cohort of 148 patients was examined, with plasma samples collected prior to treatment initiation and at the end of the first and second treatment cycles. Concentrations of 48 plasma proteins were measured using a multiplex immunoassay. The results revealed a general increase in cytokine levels following the first ipilimumab dose, consistent with immune activation. Patients not responding to treatment exhibited significantly elevated baseline levels of G-CSF, IL-2RA, MIP-1a, and SCF, compared to tumor responders (p < 0.05). Furthermore, high levels of IL-2RA, IFNγ, PDGF-bb and MIG were linked to inferior OS, while high concentrations of MIF and RANTES were associated with improved OS (p < 0.05). A multivariate model containing CRP, LDH, ECOG, IL-2RA and PDGF-bb identified a subgroup of patients with poor OS. Patients who experienced severe immune-related adverse events within three months of treatment initiation had higher baseline concentrations of several cytokines, indicating a potential association between preexisting inflammation and adverse events. These findings indicate that the first dose of ipilimumab induces a systemic response with increased levels of circulating cytokines and suggest candidate biomarkers for clinical response, immune-mediated toxicity and survival. Further studies in independent patient cohorts are required to confirm the findings.
Collapse
Affiliation(s)
| | - Claire Dunn
- Department of Cancer Immunology, Oslo University Hospital, Oslo, Norway
| | - Elin Aamdal
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Oddbjørn Straume
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tormod Kyrre Guren
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
2
|
Song X, Chen S, Cheng J, Li H, Wu R, Yan M, Wang M, Li J, Jin A, Wang W. Screening and identification Hub genes associated with immune cell infiltration and critical biomarkers in osteosarcoma. Mol Cell Probes 2025:102031. [PMID: 40374042 DOI: 10.1016/j.mcp.2025.102031] [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: 01/30/2025] [Revised: 05/03/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
PURPOSE Osteosarcoma (OS) exhibits limited immune cell infiltration that directly contributes to poor prognosis. This study sought to screen and identify pivotal biomarkers of OS immune infiltration and early diagnosis of OS. METHODS The immune cell infiltration profiles with transcriptome sequencing data from 88 OS samples were explored with CIBERSORT algorithm. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Protein-protein interaction (PPI) network analyses were applied to identify hub genes, with the expressions confirmed by dual immunofluorescence in 50 OS samples. The new biomarker gene HTRA1 were examined by immunohistochemistry and validated by the Immune score and immune gene expression profile analyses. The impact of HTRA1 on OS prognosis was verified by Least absolute shrinkage and selection operator (LASSO) regression analysis. The biological effect of HTRA1 was characterized in MG63 cells. RESULT CD8+ T cells, activated memory CD4+ T cells and plasma cells were positively correlated with the prognosis of OS. Hub genes CCL5, CXCL9, CXCL13, and HTRA1, exhibited positive correlation with the infiltration of both CD8+ T cells and CD4+ T cells. HTRA1 expression was reduced in osteosarcoma tissues, which was positively correlated with immune scores and the expressions of immune-related genes. High levels of HTRA1 were associated with favorable OS prognosis, and could negatively impacted MG63 malignant characteristics. CONCLUSION CCL5, CXCL9, CXCL13, and HTRA1 were OS hub genes positively correlate with CD8+ T cell and CD4+ T cell infiltrations. HTRA1 can serve as an underlying biomarker for the prognosis and immunotherapy of OS.
Collapse
Affiliation(s)
- Xin Song
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China; Department of Orthopedics, Xinqiao Hospital, Army Medical University.Chongqing,400038, China
| | - Sihao Chen
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| | - Junning Cheng
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou,311399, China
| | - Haiyu Li
- Institute of Intelligent Chinese Medicine,Chongqing University of Chinese Medicine,Chongqing 402760, China
| | - Ruixin Wu
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| | - Min Yan
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| | - Min Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University.Chongqing,400038, China
| | - Jie Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University.Chongqing,400038, China
| | - Aishun Jin
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| | - Wang Wang
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| |
Collapse
|
3
|
Ghanaati S, Dohle E, Schick F, Lechner J. Quantitative Real-Time RT-PCR Verifying Gene Expression Profile of Cavitations Within Human Jaw Bone. Biomedicines 2025; 13:1144. [PMID: 40426971 PMCID: PMC12109557 DOI: 10.3390/biomedicines13051144] [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: 03/03/2025] [Revised: 04/09/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Immune cells are integral to bone homeostasis, including the repair and remodeling of bone tissue. Chronic dysregulation within this osteoimmune network can lead to bone marrow defects of the jaw (BMDJ), particularly fatty degenerative osteonecrosis of the jaw (FDOJ). These localized pathologies are implicated in systemic immune dysfunctions. Methods: This study is designed to determine whether BMDJ/FDOJ samples are indicative of medullary bone pathology by evaluating FDOJ gene expression patterns using quantitative real-time PCR. Results: Comparative analyses between pathological and healthy samples evaluated the dysregulation of key molecular pathways. BMDJ/FDOJ samples showed significant upregulation of inflammatory mediators, including CCL5/RANTES, VEGF, IGF and KOR, and downregulation of structural proteins, such as collagen types I, II and IV, and osteogenesis-associated factors, such as SP7. Conclusions: The study provides new insights into the molecular mechanisms of BMDJ/FDOJ by identifying potential molecular changes suggesting a pro-inflammatory state in the affected jawbone which may contribute to systemic immune dysregulation. The findings are consistent with morphologic observations of BMDJ/FDOJ in degenerated jawbone and underscore the need for integrative approaches in dentistry and medicine while highlighting BMDJ/FDOJ as a potential target for therapeutic and preventive strategies against systemic diseases and emphasizing its clinical significance.
Collapse
Affiliation(s)
- Shahram Ghanaati
- FORM, Frankfurt Orofacial Regenerative Medicine, Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Medical Center of the Johann Wolfgang Goethe University, 60590 Frankfurt, Germany;
- ABIS e.V. (Academy for Biological Innovations in Surgery Formally Known as SBCB e.V.), Society for Blood Concentrate and Biomaterials e.V., 60435 Frankfurt, Germany
| | - Eva Dohle
- FORM, Frankfurt Orofacial Regenerative Medicine, Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Medical Center of the Johann Wolfgang Goethe University, 60590 Frankfurt, Germany;
| | - Fabian Schick
- Clinic for Integrative Dentistry, 81547 Munich, Germany; (F.S.); (J.L.)
| | - Johann Lechner
- Clinic for Integrative Dentistry, 81547 Munich, Germany; (F.S.); (J.L.)
| |
Collapse
|
4
|
Dong Z, Wu J, He L, Xie C, Geng S, Wu J, Ji X, Chen X, Zhong C, Li X. EGCG inhibits tobacco smoke-promoted proliferation of lung cancer cells through targeting CCL5. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156512. [PMID: 40010030 DOI: 10.1016/j.phymed.2025.156512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/14/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND The occurrence and development of cancer are deeply intertwined with chronic inflammatory processes. Epigallocatechin gallate (EGCG), the most pharmacologically potent catechin derived from tea, has garnered attention for its anti-inflammatory and anti-carcinogenic properties. However, the molecular mechanisms through which EGCG modulates tobacco smoke (TS)-induced inflammatory responses in lung carcinogenesis remain incompletely elucidated. PURPOSE To unravel the molecular mechanisms by which EGCG mitigates TS-induced inflammatory processes in lung carcinogenesis. METHODS Network pharmacology analysis was conducted to explore the potential target genes of EGCG involved in the inhibition of TS-induced lung cancer inflammation. In vitro and in vivo experiments were conducted to demonstrate EGCG's chemopreventive potential against lung carcinogenesis. RESULTS Utilizing data from the US adults, it was uncovered that tea consumption could suppress the inflammatory response in patients with various cancer types. CCL5 (chemokine (CC motif) ligand 5) could function as a core regulator of TS-induced lung cancer cell proliferation, and EGCG exerted beneficial effects. The following experiments revealed that TS upregulated CCL5 expression in H1299 and H226 cells. CCL5 recombinant protein elevated both ROS production and Nrf2 expression to promote lung cancer cell proliferation. EGCG could suppress CCL5-stimulated lung cancer cell proliferation by downregulating Nrf2 expression. In the mouse model, EGCG reduced tumor weight and volume, diminished the levels of CCL5, Ki67, Cyclin D1, PCNA, and Nrf2, and elevated the expression of Keap1 relative to the control group. CONCLUSION EGCG targets CCL5 to inhibit the proliferation of TS-induced lung cancer cells and may serve as a new treatment strategy.
Collapse
Affiliation(s)
- Zhiyuan Dong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China
| | - Jinyi Wu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China
| | - Liping He
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China
| | - Shanshan Geng
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China
| | - Jieshu Wu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China
| | - Xiaoming Ji
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China
| | - Xiong Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, PR China.
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China.
| | - Xiaoting Li
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, PR China.
| |
Collapse
|
5
|
Tariq MH, Advani D, Almansoori BM, AlSamahi ME, Aldhaheri MF, Alkaabi SE, Mousa M, Kohli N. The Identification of Novel Therapeutic Biomarkers in Rheumatoid Arthritis: A Combined Bioinformatics and Integrated Multi-Omics Approach. Int J Mol Sci 2025; 26:2757. [PMID: 40141401 PMCID: PMC11943070 DOI: 10.3390/ijms26062757] [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: 10/05/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 03/28/2025] Open
Abstract
Rheumatoid arthritis (RA) is a multifaceted autoimmune disease that is marked by a complex molecular profile influenced by an array of factors, including genetic, epigenetic, and environmental elements. Despite significant advancements in research, the precise etiology of RA remains elusive, presenting challenges in developing innovative therapeutic markers. This study takes an integrated multi-omics approach to uncover novel therapeutic markers for RA. By analyzing both transcriptomics and epigenomics datasets, we identified common gene candidates that span these two omics levels in patients diagnosed with RA. Remarkably, we discovered eighteen multi-evidence genes (MEGs) that are prevalent across transcriptomics and epigenomics, twelve of which have not been previously linked directly to RA. The bioinformatics analyses of the twelve novel MEGs revealed they are part of tightly interconnected protein-protein interaction networks directly related to RA-associated KEGG pathways and gene ontology terms. Furthermore, these novel MEGs exhibited direct interactions with miRNAs linked to RA, underscoring their critical role in the disease's pathogenicity. Overall, this comprehensive bioinformatics approach opens avenues for identifying new candidate markers for RA, empowering researchers to validate these markers efficiently through experimental studies. By advancing our understanding of RA, we can pave the way for more effective therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Muhammad Hamza Tariq
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Dia Advani
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates
| | - Buttia Mohamed Almansoori
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Maithah Ebraheim AlSamahi
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Maitha Faisal Aldhaheri
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Shahad Edyen Alkaabi
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Mira Mousa
- Department of Public Health and Epidemiology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates;
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Nupur Kohli
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
- Healthcare Engineering Innovation Group, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
6
|
Alrasheed AR, Awadalla M, Alnajran H, Alammash MH, Almaqati AM, Qadri I, Alosaimi B. Harnessing immunotherapeutic molecules and diagnostic biomarkers as human-derived adjuvants for MERS-CoV vaccine development. Front Immunol 2025; 16:1538301. [PMID: 40181980 PMCID: PMC11965926 DOI: 10.3389/fimmu.2025.1538301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
The pandemic potential of the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) highlights the critical need for effective vaccines due to its high fatality rate of around 36%. In this review, we identified a variety of immunotherapeutic molecules and diagnostic biomarkers that could be used in MERS vaccine development as human-derived adjuvants. We identified immune molecules that have been incorporated into standard clinical diagnostics such as CXCL10/IP10, CXCL8/IL-8, CCL5/RANTES, IL-6, and the complement proteins Ca3 and Ca5. Utilization of different human monoclonal antibodies in the treatment of MERS-CoV patients demonstrates promising outcomes in combatting MERS-CoV infections in vivo, such as hMS-1, 4C2H, 3B11-N, NBMS10-FC, HR2P-M2, SAB-301, M336, LCA60, REGN3051, REGN3048, MCA1, MERs-4, MERs-27, MERs-gd27, and MERs-gd33. Host-derived adjuvants such as CCL28, CCL27, RANTES, TCA3, and GM-CSF have shown significant improvements in immune responses, underscoring their potential to bolster both systemic and mucosal immunity. In conclusion, we believe that host-derived adjuvants like HBD-2, CD40L, and LL-37 offer significant advantages over synthetic options in vaccine development, underscoring the need for clinical trials to validate their efficacy.
Collapse
Affiliation(s)
- Abdullah R. Alrasheed
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maaweya Awadalla
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh, Saudi Arabia
| | - Hadeel Alnajran
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | | | - Adil M. Almaqati
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Ishtiaq Qadri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bandar Alosaimi
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Mirra P, Parascandolo A, Marino G, D'Alterio F, Zinna L, Desiderio A, Patitucci G, Vita GAC, Condelli V, Russi S, D'Andrea F, Beguinot F, Miele C, Formisano P, D'Esposito V. Increased levels of versican and insulin-like growth factor 1 in peritumoral mammary adipose tissue are related to aggressiveness in estrogen receptor-positive breast cancer. Mol Med 2024; 30:201. [PMID: 39501160 PMCID: PMC11539550 DOI: 10.1186/s10020-024-00968-8] [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: 05/13/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
The adipose tissue (AT) surrounding breast cancer (BC) plays a pivotal role in cancer progression and represents an optimal source for new biomarker discovery. The aim of this work was to investigate whether specific AT factors may have prognostic value in estrogen receptor-positive (ER+) BC. Proteoglycan Versican (VCAN), Insulin-like Growth Factor 1 (IGF1), Reticulon 4B (RTN4), chemokines CCL5 (also known as RANTES) and interleukin 8 (IL-8) are expressed in AT and may play important roles in BC progression. Peritumoral AT and tumoral biopsies were obtained from patients with ER+ BC (N = 23). AT specimens were collected also from healthy women (N = 17; CTRL-AT). The analysis of gene expression by qPCR revealed significantly higher mRNA levels of VCAN, IGF1, RTN4, and CCL5 in BC-AT compared to the CTRL-AT, and no difference in IL-8 mRNA levels. VCAN positively correlated with patient Body Mass Index (BMI) in BC-AT, while not in CTRL-AT. Moreover, VCAN and IGF1 positively correlated with RTN4 and negatively with CCL5. Interestingly, VCAN correlated with tumoral Ki67, while IGF1 with tumoral OCT4 that, in turn, correlated with tumoral Ki67 and patient BMI. Thus, peritumoral AT content of VCAN, and IGF1 are related to BC proliferation and aggressiveness.
Collapse
Affiliation(s)
- Paola Mirra
- URT "Genomic of Diabetes", Institute for Experimental Endocrinology and Oncology "G. Salvatore", National Research Council (IEOS-CNR), Via Pansini 5, 80131, Naples, Italy
| | - Alessia Parascandolo
- Department of Translational Medicine, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - Graziella Marino
- Department of Breast Surgery, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, Italy
| | - Federica D'Alterio
- Department of Translational Medicine, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - Lorenza Zinna
- Department of Translational Medicine, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - Antonella Desiderio
- Department of Translational Medicine, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - Giuseppe Patitucci
- Department of Anatomical Pathology, Centro di Riferimento oncologico della Basilicata (IRCCS CROB), Rionero in Vulture, Italy
| | - Giulia Anna Carmen Vita
- Department of Anatomical Pathology, Centro di Riferimento oncologico della Basilicata (IRCCS CROB), Rionero in Vulture, Italy
| | - Valentina Condelli
- Laboratory of Preclinical and Translational Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, Italy
| | - Sabino Russi
- Laboratory of Preclinical and Translational Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) CROB Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, Italy
| | - Francesco D'Andrea
- Department of Public Health, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - Francesco Beguinot
- URT "Genomic of Diabetes", Institute for Experimental Endocrinology and Oncology "G. Salvatore", National Research Council (IEOS-CNR), Via Pansini 5, 80131, Naples, Italy
- Department of Translational Medicine, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - Claudia Miele
- URT "Genomic of Diabetes", Institute for Experimental Endocrinology and Oncology "G. Salvatore", National Research Council (IEOS-CNR), Via Pansini 5, 80131, Naples, Italy
- Department of Translational Medicine, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| | - Pietro Formisano
- URT "Genomic of Diabetes", Institute for Experimental Endocrinology and Oncology "G. Salvatore", National Research Council (IEOS-CNR), Via Pansini 5, 80131, Naples, Italy.
- Department of Translational Medicine, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy.
| | - Vittoria D'Esposito
- URT "Genomic of Diabetes", Institute for Experimental Endocrinology and Oncology "G. Salvatore", National Research Council (IEOS-CNR), Via Pansini 5, 80131, Naples, Italy
- Department of Translational Medicine, University of Naples "Federico II", Via Pansini 5, 80131, Naples, Italy
| |
Collapse
|
8
|
Guan Y, Liu X, Tian J, Yang G, Xu F, Guo N, Guo L, Wan Z, Huang Z, Gao M, Chong T. CCL5 promotes the epithelial-mesenchymal transition of circulating tumor cells in renal cancer. J Transl Med 2024; 22:817. [PMID: 39227943 PMCID: PMC11370314 DOI: 10.1186/s12967-024-05297-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/12/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are pivotal in tumor metastasis across cancers, yet their specific role in renal cancer remains unclear. METHODS This study investigated C-C motif chemokine ligand 5 (CCL5)'s tumorigenic impact on renal cancer cells and CTCs using bioinformatics, in vivo, and in vitro experiments. It also assessed renal cancer patients' CTCs prognostic value through Lasso regression and Kaplan-Meier survival curves. RESULTS Bioinformatics analysis revealed differential genes focusing on cellular adhesion and migration between CTCs and tumor cells. CCL5 exhibited high expression in various CTCs, correlating with poor prognosis in renal cancer. In 786-O-CTCs, CCL5 enhanced malignancy, while in renal cell carcinoma cell line CAKI-2 and 786-O, it promoted epithelial-mesenchymal transition (EMT) via smad2/3, influencing cellular characteristics. The nude mouse model suggested CCL5 increased CTCs and intensified EMT, enhancing lung metastasis. Clinical results shown varying prognostic values for different EMT-typed CTCs, with mesenchymal CTCs having the highest value. CONCLUSIONS In summary, CCL5 promoted EMT in renal cancer cells and CTCs through smad2/3, enhancing the malignant phenotype and facilitating lung metastasis. Mesenchymal-type CTC-related factors can construct a risk model for renal cancer patients, allowing personalized treatment based on metastatic risk prediction.
Collapse
Affiliation(s)
- Yibing Guan
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
- Department of Urology, The First Affiliated Hospital, Zhengzhou University, No 1 Jianshe East Road, Zhengzhou, 450052, He Nan, China
| | - Xueyi Liu
- School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Juanhua Tian
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Guang Yang
- Henan Key Lab Reprod & Genet, The First Affiliated Hospital, Zhengzhou University, No 1 Jianshe East Road, Zhengzhou, 450052, He Nan, China
| | - Fangshi Xu
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Ni Guo
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Lingyu Guo
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Ziyan Wan
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Zhixin Huang
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Mei Gao
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Tie Chong
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China.
| |
Collapse
|
9
|
Zhang C, Sun N, Fei Q, Peng L, Wei C, Liu X, Miao S, Chai M, Wang F, Wang D, Hong J, Huang S, Zhang S, Qiu H. MEN1 deficiency stabilizes PD-L1 and promotes tumor immune evasion of lung cancer. Cancer Sci 2024; 115:2515-2527. [PMID: 38685894 PMCID: PMC11309931 DOI: 10.1111/cas.16196] [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: 12/10/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
Multiple Endocrine Neoplasia 1 gene (MEN1), which is known to be a tumor suppressor gene in lung tissues, encodes a 610 amino acid protein menin. Previous research has proven that MEN1 deficiency promotes the malignant progression of lung cancer. However, the biological role of this gene in the immune microenvironment of lung cancer remains unclear. In this study, we found that programmed cell death-ligand 1 (PD-L1) is upregulated in lung-specific KrasG12D mutation-induced lung adenocarcinoma in mice, after Men1 deficiency. Simultaneously, CD8+ and CD3+ T cells are depleted, and their cytotoxic effects are suppressed. In vitro, PD-L1 is inhibited by the overexpression of menin. Mechanistically, we found that MEN1 inactivation promotes the deubiquitinating activity of COP9 signalosome subunit 5 (CSN5) and subsequently increases the level of PD-L1.
Collapse
Affiliation(s)
- Cuncun Zhang
- School of NursingAnhui Medical UniversityHefeiChina
| | - Ningning Sun
- School of NursingAnhui Medical UniversityHefeiChina
| | - Qingze Fei
- School of NursingAnhui Medical UniversityHefeiChina
| | - Linlin Peng
- School of NursingAnhui Medical UniversityHefeiChina
| | - Chengyu Wei
- School of NursingAnhui Medical UniversityHefeiChina
| | - Xiangyu Liu
- School of NursingAnhui Medical UniversityHefeiChina
| | - Sainan Miao
- School of NursingAnhui Medical UniversityHefeiChina
| | - Mengqi Chai
- School of NursingAnhui Medical UniversityHefeiChina
| | - Fang Wang
- Department of PathologyZhejiang HospitalHangzhouChina
| | - Di Wang
- School of NursingAnhui Medical UniversityHefeiChina
| | | | - Shenghai Huang
- Department of Microbiology, The Institute of Clinical Virology, School of Basic Medical Sciences, Anhui Medical UniversityHefeiChina
| | - Shihao Zhang
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of EducationAnhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiChina
| | - Huan Qiu
- School of NursingAnhui Medical UniversityHefeiChina
| |
Collapse
|
10
|
Simmen FA, Alhallak I, Simmen RCM. Krüppel-like Factor-9 and Krüppel-like Factor-13: Highly Related, Multi-Functional, Transcriptional Repressors and Activators of Oncogenesis. Cancers (Basel) 2023; 15:5667. [PMID: 38067370 PMCID: PMC10705314 DOI: 10.3390/cancers15235667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 02/12/2024] Open
Abstract
Specificity Proteins/Krüppel-like Factors (SP/KLF family) are a conserved family of transcriptional regulators. These proteins share three highly conserved, contiguous zinc fingers in their carboxy-terminus, requisite for binding to cis elements in DNA. Each SP/KLF protein has unique primary sequence within its amino-terminal and carboxy-terminal regions, and it is these regions which interact with co-activators, co-repressors, and chromatin-modifying proteins to support the transcriptional activation and repression of target genes. Krüppel-like Factor 9 (KLF9) and Krüppel-like Factor 13 (KLF13) are two of the smallest members of the SP/KLF family, are paralogous, emerged early in metazoan evolution, and are highly conserved. Paradoxically, while most similar in primary sequence, KLF9 and KLF13 display many distinct roles in target cells. In this article, we summarize the work that has identified the roles of KLF9 (and to a lesser degree KLF13) in tumor suppression or promotion via unique effects on differentiation, pro- and anti-inflammatory pathways, oxidative stress, and tumor immune cell infiltration. We also highlight the great diversity of miRNAs, lncRNAs, and circular RNAs which provide mechanisms for the ubiquitous tumor-specific suppression of KLF9 mRNA and protein. Elucidation of KLF9 and KLF13 in cancer biology is likely to provide new inroads to the understanding of oncogenesis and its prevention and treatments.
Collapse
Affiliation(s)
- Frank A. Simmen
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (I.A.); (R.C.M.S.)
- The Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Iad Alhallak
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (I.A.); (R.C.M.S.)
| | - Rosalia C. M. Simmen
- Department of Physiology & Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (I.A.); (R.C.M.S.)
- The Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|