2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Single-cell sequencing analysis related to sphingolipid metabolism guides immunotherapy and prognosis of skin cutaneous melanoma

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          We explore sphingolipid-related genes (SRGs) in skin melanoma (SKCM) to develop a prognostic indicator for patient outcomes. Dysregulated lipid metabolism is linked to aggressive behavior in various cancers, including SKCM. However, the exact role and mechanism of sphingolipid metabolism in melanoma remain partially understood.

          Methods

          We integrated scRNA-seq data from melanoma patients sourced from the GEO database. Through the utilization of the Seurat R package, we successfully identified distinct gene clusters associated with patient survival in the scRNA-seq data. Key prognostic genes were identified through single-factor Cox analysis and used to develop a prognostic model using LASSO and stepwise regression algorithms. Additionally, we evaluated the predictive potential of these genes within the immune microenvironment and their relevance to immunotherapy. Finally, we validated the functional significance of the high-risk gene IRX3 through in vitro experiments.

          Results

          Analysis of scRNA-seq data identified distinct expression patterns of 4 specific genes (SRGs) in diverse cell subpopulations. Re-clustering cells based on increased SRG expression revealed 7 subgroups with significant prognostic implications. Using marker genes, lasso, and Cox regression, we selected 11 genes to construct a risk signature. This signature demonstrated a strong correlation with immune cell infiltration and stromal scores, highlighting its relevance in the tumor microenvironment. Functional studies involving IRX3 knockdown in A375 and WM-115 cells showed significant reductions in cell viability, proliferation, and invasiveness.

          Conclusion

          SRG-based risk signature holds promise for precise melanoma prognosis. An in-depth exploration of SRG characteristics offers insights into immunotherapy response. Therapeutic targeting of the IRX3 gene may benefit melanoma patients.

          Related collections

          Most cited references40

          • Record: found
          • Abstract: found
          • Article: not found

          Microenvironmental regulation of tumor progression and metastasis.

          Cancers develop in complex tissue environments, which they depend on for sustained growth, invasion and metastasis. Unlike tumor cells, stromal cell types within the tumor microenvironment (TME) are genetically stable and thus represent an attractive therapeutic target with reduced risk of resistance and tumor recurrence. However, specifically disrupting the pro-tumorigenic TME is a challenging undertaking, as the TME has diverse capacities to induce both beneficial and adverse consequences for tumorigenesis. Furthermore, many studies have shown that the microenvironment is capable of normalizing tumor cells, suggesting that re-education of stromal cells, rather than targeted ablation per se, may be an effective strategy for treating cancer. Here we discuss the paradoxical roles of the TME during specific stages of cancer progression and metastasis, as well as recent therapeutic attempts to re-educate stromal cells within the TME to have anti-tumorigenic effects.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Genetic basis for clinical response to CTLA-4 blockade in melanoma.

            Immune checkpoint inhibitors are effective cancer treatments, but molecular determinants of clinical benefit are unknown. Ipilimumab and tremelimumab are antibodies against cytotoxic T-lymphocyte antigen 4 (CTLA-4). Anti-CTLA-4 treatment prolongs overall survival in patients with melanoma. CTLA-4 blockade activates T cells and enables them to destroy tumor cells. We obtained tumor tissue from patients with melanoma who were treated with ipilimumab or tremelimumab. Whole-exome sequencing was performed on tumors and matched blood samples. Somatic mutations and candidate neoantigens generated from these mutations were characterized. Neoantigen peptides were tested for the ability to activate lymphocytes from ipilimumab-treated patients. Malignant melanoma exomes from 64 patients treated with CTLA-4 blockade were characterized with the use of massively parallel sequencing. A discovery set consisted of 11 patients who derived a long-term clinical benefit and 14 patients who derived a minimal benefit or no benefit. Mutational load was associated with the degree of clinical benefit (P=0.01) but alone was not sufficient to predict benefit. Using genomewide somatic neoepitope analysis and patient-specific HLA typing, we identified candidate tumor neoantigens for each patient. We elucidated a neoantigen landscape that is specifically present in tumors with a strong response to CTLA-4 blockade. We validated this signature in a second set of 39 patients with melanoma who were treated with anti-CTLA-4 antibodies. Predicted neoantigens activated T cells from the patients treated with ipilimumab. These findings define a genetic basis for benefit from CTLA-4 blockade in melanoma and provide a rationale for examining exomes of patients for whom anti-CTLA-4 agents are being considered. (Funded by the Frederick Adler Fund and others.).
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Sphingolipids and their metabolism in physiology and disease

              Studies of bioactive lipids in general and sphingolipids in particular have intensified over the past several years, revealing an unprecedented and unanticipated complexity of the lipidome and its many functions, which rivals, if not exceeds, that of the genome or proteome. These results highlight critical roles for bioactive sphingolipids in most, if not all, major cell biological responses, including all major cell signalling pathways, and they link sphingolipid metabolism to key human diseases. Nevertheless, the fairly nascent field of bioactive sphingolipids still faces challenges in its biochemical and molecular underpinnings, including defining the molecular mechanisms of pathway and enzyme regulation, the study of lipid-protein interactions and the development of cellular probes, suitable biomarkers and therapeutic approaches.
                Bookmark

                Author and article information

                Contributors
                URI : https://loop.frontiersin.org/people/2358849Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1013714Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/2144186Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1369836Role: Role: Role: Role:
                Role: Role: Role:
                Role: Role: Role:
                Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1443376Role: Role: Role:
                Role: Role: Role:
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                23 November 2023
                2023
                : 14
                : 1304466
                Affiliations
                [1] 1 Department of Dermatology, The First Affiliated Hospital of Anhui Medical University , Hefei, Anhui, China
                [2] 2 Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education , Hefei, Anhui, China
                [3] 3 Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province , Hefei, Anhui, China
                [4] 4 Department of Plastic Surgery, The Ninth Affiliated Hospital of Shanghai Jiaotong University , Shanghai, China
                [5] 5 Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University , Hefei, China
                [6] 6 Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University , Hefei, Anhui, China
                [7] 7 Department of Plastic Surgery, The First Affiliated Hospital of Anhui Medical University , Hefei, Anhui, China
                [8] 8 Department of Gynecology, The First Affiliated Hospital of Anhui Medical University , Hefei, Anhui, China
                Author notes

                Edited by: Chen Yang, German Cancer Research Center (DKFZ), Germany

                Reviewed by: Yunfei Liu, Ludwig Maximilian University of Munich, Germany; Kangjie Shen, Fudan University, China; Zhou Sun, Jilin University, China

                *Correspondence: Shengxiu Liu, liushengxiu@ 123456ahmu.edu.cn ; Wenming Zhou, aydzwm@ 123456163.com
                Article
                10.3389/fimmu.2023.1304466
                10701528
                38077400
                0c65ea03-f165-451a-8576-6f17a34f1ec4
                Copyright © 2023 Ding, Zhao, Cai, Zhou, Chen, Bai, Liu, Liu and Zhou

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

                History
                : 29 September 2023
                : 07 November 2023
                Page count
                Figures: 10, Tables: 0, Equations: 0, References: 40, Pages: 18, Words: 6781
                Funding
                The author(s) declare financial support was received for the research, authorship, and/or publication of this article. The present study was financially supported by the Scientific research project of universities in Anhui Province (grant nos. 2022AH040163), and the Postgraduate Innovation Research and Practice Program of Anhui Medical University (grant nos. YJS20230068 and YJS20230074).
                Categories
                Immunology
                Original Research
                Custom metadata
                Cancer Immunity and Immunotherapy

                Immunology
                melanoma,sphingolipid,irx3,single-cell sequencing,immunotherapy
                Immunology
                melanoma, sphingolipid, irx3, single-cell sequencing, immunotherapy

                Comments

                Comment on this article