1
|
Zhou Y, Kou J, Li W, Wang Y, Su X, Zhang H. BCAA metabolism in cancer progression and therapy resistance: The balance between fuel and cell signaling. Front Pharmacol 2025; 16:1595176. [PMID: 40438606 PMCID: PMC12116492 DOI: 10.3389/fphar.2025.1595176] [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: 03/17/2025] [Accepted: 05/01/2025] [Indexed: 06/01/2025] Open
Abstract
Branched-chain amino acids (BCAAs), including leucine, isoleucine, and valine, play a crucial role in cellular metabolism and signaling. Recent studies have demonstrated that BCAA metabolic reprogramming is a key driver of tumor progression and treatment resistance in various cancers. BCAA metabolism supports cancer cell growth, survival, and proliferation by modulating pathways such as mTOR signaling and oxidative stress responses. By promoting immunosuppressive conditions and increasing the survival rate of cancer stem cells (CSCs), BCAAs contribute to immune evasion and resistance to therapies such as chemotherapy and immune checkpoint inhibitors. This article explores the different metabolic reprogramming patterns of BCAAs in various tumors and introduces BCAA-related metabolic targets for overcoming tumor resistance, offering new directions for precision cancer treatment, reducing resistance, and improving patient outcomes.
Collapse
Affiliation(s)
- Yi Zhou
- Departments of Thoracic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiahui Kou
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Wenjin Li
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Yuyao Wang
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Xingxing Su
- Shunyi Maternal and Children’s Hospital of Beijing Children’s Hospital, Beijing, China
| | - Hongguang Zhang
- Departments of Thoracic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Yi X, Tang B, Mo Q, Tang Y, Fu W, Zhang L, Xie L. Identification of Immune Characteristics of 2 Subtypes of Breast Cancer by Combining Polyamine Metabolism-related Genes to Help With Immunotherapy. J Immunother 2025:00002371-990000000-00139. [PMID: 40302111 DOI: 10.1097/cji.0000000000000559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/20/2025] [Indexed: 05/01/2025]
Abstract
This project aims to explore the clustering value of polyamine metabolism-related genes (PMRGs) in breast cancer (BC) to assist treatment. ConsensusClusterPlus R package was employed to cluster BC patients based on the expression of PMRGs. Using the edgeR R package, we analyzed differentially expressed genes (DEGs) of different molecular clusters. Core genes were screened and enriched by the PPI network. Univariate COX was applied to determine genes tightly linked with survival. ConsensusClusterPlus R package was employed to cluster PMRGs. Differences in immune infiltration and expression of immune checkpoints between 2 subgroups were analyzed. Response to immunotherapy was assessed based on the expression level of immunophenoscore (IPS). Drug sensitivity of different PMRG clusters was assessed by pRRophitic R package. We clustered BC patients into 2 different subtypes with different survival rates and biological functions based on the expression of 16 PMRGs. Application of univariate COX analysis identified genes greatly associated with survival and divided BC patients into 2 different PMRG clusters. Patients in the 2 clusters exhibited differences in overall survival rate and immune cell infiltration levels, with multiple immune cells displaying higher immune levels in PMRG cluster 2. PMRG cluster 2 demonstrated higher expression of HLA and IC as well as IPS. Cluster 1 exhibited higher sensitivity to (5Z)-7-Oxozeaenol, 5-Fluorouracil, and 681640, while cluster 2 exhibited higher sensitivity to A-443654 and A-770041. We identified 2 clusters of PMRG with significant differences in the immune microenvironment in BC and predicted potential drugs, aiming to find new directions for clinical treatment of BC.
Collapse
Affiliation(s)
- Xiuwen Yi
- Department of Medical Oncology, Hengyang Medical School
- Department of Medical Oncology, The First Affiliated Hospital of University of South China
| | | | | | | | - Wei Fu
- Department of Orthopedics, The First People's Hospital of Hengyang, Hengyang
| | - Lingling Zhang
- School of Medical Health Management, Hunan Vocational College of Foreign Languages, Changsha, Hunan, China
| | - Liming Xie
- Department of Medical Oncology, The First Affiliated Hospital of University of South China
| |
Collapse
|
3
|
Yu Y, Liu H, Liu K, Zhao M, Zhang Y, Jiang R, Wang F. Multi-omics identification of a polyamine metabolism related signature for hepatocellular carcinoma and revealing tumor microenvironment characteristics. Front Immunol 2025; 16:1570378. [PMID: 40330470 PMCID: PMC12052762 DOI: 10.3389/fimmu.2025.1570378] [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: 02/03/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Background Accumulating evidence indicates that elevated polyamine levels are closely linked to tumor initiation and progression. However, the precise role of polyamine metabolism in hepatocellular carcinoma (HCC) remains poorly understood. Methods We conducted differential expression analysis on bulk RNA sequencing data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) to identify 65 polyamine metabolism-related genes. By employing unsupervised consensus clustering, AddModuleScore, single-sample gene set enrichment analysis (ssGSEA), and weighted gene co-expression network analysis (WGCNA), we identified polyamine metabolism-related genes at both the bulk RNA-seq and single-cell RNA-seq (scRNA-seq) levels. Utilizing 101 machine learning algorithms, we constructed a polyamine metabolism-related signature (PMRS) and validated its predictive power across training, testing, and external validation cohorts. Additionally, we developed a prognostic nomogram model by integrating PMRS with clinical variables. To explore immune treatment sensitivity, we assessed tumor mutation burden (TMB), tumor immune dysfunction and exclusion (TIDE) score, mutation frequency, and immune checkpoint genes expression. Immune cell infiltration was analyzed using the CIBERSORT algorithm. Finally, RT-qPCR experiments were conducted to validate the expression of key genes. Results Using 101 machine learning algorithms, we established a polyamine metabolism-related signature comprising 9 genes, which exhibited strong prognostic value for HCC patients. Further analysis revealed significant differences in clinical features, biological functions, mutation profiles, and immune cell infiltration between high-risk and low-risk groups. Notably, TIDE analysis and immune phenotype scoring (IPS) demonstrated distinct immune treatment sensitivities between the two risk groups. RT-qPCR validation confirmed that these 9 genes were highly expressed in normal cells but significantly downregulated in tumor cells. Conclusions Our study developed a polyamine metabolism-based prognostic risk signature for HCC, which may provide valuable insights for personalized treatment strategies in HCC patients.
Collapse
Affiliation(s)
- Yuexi Yu
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| | - Huiru Liu
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| | - Kaipeng Liu
- Department of Hepatobiliary Oncology, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Meiqi Zhao
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Nankai University, Tianjin, China
| | - Yiyan Zhang
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| | - Runci Jiang
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| | - Fengmei Wang
- Department of gastroenterology &hepatology, Tianjin First Center Hospital, Tianjin Key Laboratory for Organ Transplantation, Tianjin Key Laboratory of Molecular Diagnosis and Treatment of Liver Cancer, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Yu J, Zhang C, Zhang Q, Lu B, Lu G, Zhang C, Qiu R, Wang X, Zou C, Chu J, Li H, Zhao W. AZIN1-dependent polyamine synthesis accelerates tumor cell cycle progression and impairs effector T-cell function in osteosarcoma. Cell Death Dis 2025; 16:310. [PMID: 40246846 PMCID: PMC12006533 DOI: 10.1038/s41419-025-07640-x] [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: 10/28/2024] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025]
Abstract
Osteosarcoma, the most prevalent malignant bone tumor among adolescents, frequently exhibits limited responsiveness to immunotherapy, a challenge attributed to poorly understood underlying mechanisms. Here, we identify enhanced polyamine biosynthesis as a key driver of osteosarcoma progression and immunotherapy resistance. We show that osteosarcoma cell proliferation and tumor growth rely on polyamine availability and that disruption of polyamine synthesis significantly boosts the cytotoxic efficacy of TCR-engineered T cells against osteosarcoma cells. Mechanistically, we reveal that the knockdown of antizyme inhibitor 1 (AZIN1) or suppression of polyamine production reduces MYC expression, leading to diminished tumor cell viability via the downregulation of cell cycle-related genes. Furthermore, reduced MYC levels are associated with changes in the expression of immunomodulatory cytokines and human leukocyte antigen molecules, pointing to a potential link with enhanced T-cell-mediated cytotoxicity. Collectively, our findings establish a pivotal role for the AZIN1-polyamine axis in osteosarcoma proliferation and immune evasion, and support the development of novel immunotherapeutic strategies targeting polyamine biosynthesis to combat this aggressive cancer.
Collapse
Affiliation(s)
- Jiaming Yu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chuanxia Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Qinkai Zhang
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, China
| | - Bing Lu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Guohao Lu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510000, China
| | - Chunxiao Zhang
- School of Laboratory Medicine, Guangzhou Health Science College, Guangzhou, 510450, China
| | - Ru Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510000, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510000, China
| | - Xinyue Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510000, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510000, China
| | - Changye Zou
- Musculoskeletal Oncology Center, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Junjun Chu
- HRYZ Biotech Co., Guangzhou, 510507, China.
| | - Haizhou Li
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, 341000, China.
| | - Wei Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510000, China.
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510000, China.
| |
Collapse
|
5
|
Filisola-Villaseñor JG, Arroyo-Sánchez BI, Navarro-González LJ, Morales-Ríos E, Olin-Sandoval V. Ornithine decarboxylase and its role in cancer. Arch Biochem Biophys 2025; 765:110321. [PMID: 39870288 DOI: 10.1016/j.abb.2025.110321] [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: 08/30/2024] [Revised: 01/03/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
Cancer is among the leading causes of death worldwide. The effectiveness of conventional chemotherapy has some drawbacks, therefore, there is an urgency to develop novel strategies to fight this disease. Ornithine decarboxylase (ODC) is the most finely tuned enzyme of the polyamine (PA) biosynthesis pathway as it is regulated at different levels: transcriptional, translational, post-translational, and by feedback inhibition. In cancer, this enzyme is overexpressed due to its regulation by the protooncogene c-Myc, thus it has been proposed as a drug target against this disease. This review describes information regarding the biochemistry and regulation of the ODC at different levels and its role in cancer. Moreover, we discuss the molecules aiming on the inhibition of the ODC activity that have been tested as therapeutic options. ODC remains as a therapeutic opportunity that needs to be more explored.
Collapse
Affiliation(s)
| | - Beatriz Irene Arroyo-Sánchez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Luis Janiel Navarro-González
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Edgar Morales-Ríos
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico.
| | - Viridiana Olin-Sandoval
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico.
| |
Collapse
|
6
|
Sun Y, Zhou P, Qian J, Zeng Q, Wei G, Li Y, Liu Y, Lai Y, Zhan Y, Wu D, Fang Y. Spermine synthase engages in macrophages M2 polarization to sabotage antitumor immunity in hepatocellular carcinoma. Cell Death Differ 2025; 32:573-586. [PMID: 39658701 PMCID: PMC11894157 DOI: 10.1038/s41418-024-01409-z] [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: 03/07/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 12/12/2024] Open
Abstract
Disturbances in tumor cell metabolism reshape the tumor microenvironment (TME) and impair antitumor immunity, but the implicit mechanisms remain elusive. Here, we found that spermine synthase (SMS) was significantly upregulated in tumor cells, which correlated positively with the immunosuppressive microenvironment and predicted poor survival in hepatocellular carcinoma (HCC) patients. Via "subcutaneous" and "orthotopic" HCC syngeneic mouse models and a series of in vitro coculture experiments, we identified elevated SMS levels in HCC cells played a role in immune escape mainly through its metabolic product spermine, which induced M2 polarization of tumor-associated macrophages (TAMs) and subsequently corresponded with a decreased antitumor functionality of CD8+ T cells. Mechanistically, we discovered that spermine reprogrammed TAMs mainly by activating the PI3K-Akt-mTOR-S6K signaling pathway. Spermine inhibition in combination with immune checkpoint blockade effectively diminished tumor burden in vivo. Our results expand the understanding of the critical role of metabolites in regulating cancer progression and antitumor immunity and open new avenues for developing novel therapeutic strategies against HCC.
Collapse
Affiliation(s)
- Yining Sun
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangzhou, Guangdong Province, China
| | - Peitao Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangzhou, Guangdong Province, China
| | - Junying Qian
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qin Zeng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guangyan Wei
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yongsheng Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuechen Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yingjie Lai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yizhi Zhan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Dehua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
- Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangzhou, Guangdong Province, China.
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
- Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangzhou, Guangdong Province, China.
| |
Collapse
|
7
|
Wu JY, Zeng Y, You YY, Chen QY. Polyamine metabolism and anti-tumor immunity. Front Immunol 2025; 16:1529337. [PMID: 40040695 PMCID: PMC11876390 DOI: 10.3389/fimmu.2025.1529337] [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: 11/16/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Growing attention has been directed toward the critical role of polyamines in the tumor microenvironment and immune regulation. Polyamines, primarily comprising putrescine, spermidine, and spermine, are tightly regulated through coordinated biosynthesis, catabolism, and transport, with distinct metabolic patterns between normal and cancerous tissues. Emerging evidence highlights the pivotal role of polyamine metabolism in tumor initiation, progression, and metastasis. This review aims to elucidate the differences in polyamine biosynthesis, transport, and catabolism between normal and cancerous tissues, as well as the associated alterations in tumor epigenetic modifications and resistance to immune checkpoint blockade driven by polyamine metabolism. Polyamine metabolism influences both tumor cells and the tumor microenvironment by modulating immune cell phenotypes-shifting them towards either tumor suppression or immune evasion within the tumor immune microenvironment. Additionally, polyamine metabolism impacts immunotherapy through its regulation of key enzymes. This review also explores potential therapeutic targets and summarizes the roles of polyamine inhibitors in combination with immunotherapy for cancer treatment, offering a novel perspective on therapeutic strategies.
Collapse
Affiliation(s)
- Jing-Yi Wu
- Fujian Medical University, Fuzhou, Fujian, China
| | - Yan Zeng
- Fujian Medical University, Fuzhou, Fujian, China
| | - Yu-Yang You
- Fujian Medical University, Fuzhou, Fujian, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
8
|
Mafe AN, Büsselberg D. Modulation of the Neuro-Cancer Connection by Metabolites of Gut Microbiota. Biomolecules 2025; 15:270. [PMID: 40001573 PMCID: PMC11853082 DOI: 10.3390/biom15020270] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
The gut-brain-cancer axis represents a novel and intricate connection between the gut microbiota, neurobiology, and cancer progression. Recent advances have accentuated the significant role of gut microbiota metabolites in modulating systemic processes that influence both brain health and tumorigenesis. This paper explores the emerging concept of metabolite-mediated modulation within the gut-brain-cancer connection, focusing on key metabolites such as short-chain fatty acids (SCFAs), tryptophan derivatives, secondary bile acids, and lipopolysaccharides (LPS). While the gut microbiota's impact on immune regulation, neuroinflammation, and tumor development is well established, gaps remain in grasping how specific metabolites contribute to neuro-cancer interactions. We discuss novel metabolites with potential implications for neurobiology and cancer, such as indoles and polyamines, which have yet to be extensively studied. Furthermore, we review preclinical and clinical evidence linking gut dysbiosis, altered metabolite profiles, and brain tumors, showcasing limitations and research gaps, particularly in human longitudinal studies. Case studies investigating microbiota-based interventions, including dietary changes, fecal microbiota transplantation, and probiotics, demonstrate promise but also indicate hurdles in translating these findings to clinical cancer therapies. This paper concludes with a call for standardized multi-omics approaches and bi-directional research frameworks integrating microbiome, neuroscience, and oncology to develop personalized therapeutic strategies for neuro-cancer patients.
Collapse
Affiliation(s)
- Alice N. Mafe
- Department of Biological Sciences, Faculty of Sciences, Taraba State University, Main Campus, Jalingo 660101, Taraba State, Nigeria;
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha Metropolitan Area, Doha P.O. Box 22104, Qatar
| |
Collapse
|
9
|
Yu S, Zhao Y, Liu Q, Wang J, Fu J, Li R, Yuan Y, Yan X, Su J. Spermidine synthase promotes liver cancer progression in a paracrine manner by altering the macrophage immunometabolic state. Bioorg Chem 2025; 155:108135. [PMID: 39793221 DOI: 10.1016/j.bioorg.2025.108135] [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: 08/15/2024] [Revised: 11/03/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
PURPOSE Understanding the molecular mechanisms of adaptive regulation in the tumor microenvironment is crucial for precision therapy in hepatocellular carcinoma (HCC). We hypothesized that cargo proteins carried by extracellular vesicles (EVs) released in a hypoxic microenvironment might promote HCC progression by remodeling tumor-associated macrophages (TAMs). METHODS EV protein analysis by label-free proteomics mass spectrometry of HCC cell lines of different tumor grades was performed. The promotional effect if spermidine synthase(SRM) on M2 polarized TAMs was further investigated using various biological approaches. RESULTS SRM expression was positively correlated with liver cancer progression in HCC cell lines, liver cancer samples, and nude mouse models. In a mouse model, SRM expression was positively correlated with TAM infiltration and liver cancer progression. Pan-cancer dataset analysis confirmed that SRM overexpression in HCC tumors is correlated with poor patient prognosis. However, a hypoxic microenvironment is an internal driving factor for exosomal SRM that participates in microenvironmental modifications. Moreover, we defined a hitherto unknown pattern of microenvironmental crosstalk involving SRM in EVs, whereby macrophages complete the phenotypic fate of M2 tumor-associated macrophages through SRM uptake. CONCLUSION SRM regulation within the immune microenvironment is metabolically driven. By upregulating spermidine, which serves as a substrate for eIF5A hypusination, excessive oxidative phosphorylation (OXPHOS) assembly is achieved. This, in turn, leads to the expression of immunosuppressive marker molecules and ultimately promotes liver cancer progression. SRM, which is enriched in the EVs of HCC cells under hypoxic conditions, acts as a potent regulator linking polyamine and energy metabolism in TAMs, thereby promoting liver cancer progression.
Collapse
Affiliation(s)
- Sihang Yu
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China
| | - Yuanxin Zhao
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China
| | - Qingqing Liu
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China
| | - Jian Wang
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China
| | - Jiaying Fu
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China
| | - Runyuan Li
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China
| | - Yuan Yuan
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China
| | - Xiaoyu Yan
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China
| | - Jing Su
- Key Laboratory of Pathobiology Ministry of Education Department of Pathophysiology College of Basic Medical Sciences Jilin University Changchun China.
| |
Collapse
|
10
|
Kim MJ, Chang JH. Structure simulation-based comparison of active site variations in fungal ornithine decarboxylases. Commun Integr Biol 2025; 18:2458872. [PMID: 39906711 PMCID: PMC11792860 DOI: 10.1080/19420889.2025.2458872] [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: 07/16/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
Polyamines play crucial roles in various biological processes, including cell proliferation and differentiation, immune response modulation, and signal transduction. Ornithine decarboxylase (ODC) initiates polyamine biosynthesis by catalyzing the conversion of ornithine to putrescine in a pyridoxal phosphate (PLP)-dependent manner. While the structures of mammalian and protozoan ODCs have been elucidated, fungal ODCs remain uncharacterized. In this study, AlphaFold2 was employed to simulate the structures of ODCs from four fungi: Kluyveromyces lactis, Candida albicans, Debaryomyces hansenii, and Schizosaccharomyces pombe. The results indicated that, although these ODCs share α/β-barrel and β-sheet domains, their active site conformations exhibit subtle differences. Additionally, substrate selectivity among ODCs and related decarboxylases varied depending on the distance between the Cα of aspartate or glutamate residues within the specificity helix and the C4α of PLP. Notably, the bacterial Campylobacter jejuni decarboxylase (CjCANSDC), which binds the largest substrate, exhibits the longest distance, whereas fungal ODC, which binds the smallest substrate, displays the shortest distance. Furthermore, significant differences in the composition of amino acid residues within the active sites were also observed. This study provides insights into the structural diversity and catalytic activity of ODCs across a broad range of organisms, advancing the understanding of structure-dependent evolutionary processes.
Collapse
Affiliation(s)
- Min Jeong Kim
- Department of Biology Education, Kyungpook National University, Daegu, South Korea
| | - Jeong Ho Chang
- Department of Biology Education, Kyungpook National University, Daegu, South Korea
- Department of Biomedical Convergence Science and Technology, Kyungpook National University, Daegu, South Korea
- Science Education Research Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
11
|
Khan T, Nagarajan M, Kang I, Wu C, Wangpaichitr M. Targeting Metabolic Vulnerabilities to Combat Drug Resistance in Cancer Therapy. J Pers Med 2025; 15:50. [PMID: 39997327 PMCID: PMC11856717 DOI: 10.3390/jpm15020050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Drug resistance remains a significant barrier to effective cancer therapy. Cancer cells evade treatment by reprogramming their metabolism, switching from glycolysis to oxidative phosphorylation (OXPHOS), and relying on alternative carbon sources such as glutamine. These adaptations not only enable tumor survival but also contribute to immune evasion through mechanisms such as reactive oxygen species (ROS) generation and the upregulation of immune checkpoint molecules like PD-L1. This review explores the potential of targeting metabolic weaknesses in drug-resistant cancers to enhance therapeutic efficacy. Key metabolic pathways involved in resistance, including glycolysis, glutamine metabolism, and the kynurenine pathway, are discussed. The combination of metabolic inhibitors with immune checkpoint inhibitors (ICIs), particularly anti-PD-1/PD-L1 therapies, represents a promising approach to overcoming both metabolic and immune evasion mechanisms. Clinical trials combining metabolic and immune therapies have shown early promise, but further research is needed to optimize treatment combinations and identify biomarkers for patient selection. In conclusion, targeting cancer metabolism in combination with immune checkpoint blockade offers a novel approach to overcoming drug resistance, providing a potential pathway to improved outcomes in cancer therapy. Future directions include personalized treatments based on tumor metabolic profiles and expanding research to other tumor types.
Collapse
Affiliation(s)
- Taranatee Khan
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
| | - Manojavan Nagarajan
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Irene Kang
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
- South Florida VA Foundation for Research and Education, Miami, FL 33125, USA
| | - Chunjing Wu
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
| | - Medhi Wangpaichitr
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33125, USA; (T.K.); (M.N.); (I.K.); (C.W.)
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- South Florida VA Foundation for Research and Education, Miami, FL 33125, USA
- Department of Surgery, Division of Thoracic Surgery, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
12
|
Kim MS, Kang H, Baek JH, Cho MG, Chung EJ, Kim SJ, Chung JY, Chun KH. Disrupting Notch signaling related HES1 in myeloid cells reinvigorates antitumor T cell responses. Exp Hematol Oncol 2024; 13:122. [PMID: 39702544 DOI: 10.1186/s40164-024-00588-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are immunosuppressive cells within the tumor microenvironment (TME) that hinder anti-tumor immunity. Notch signaling is a pathway crucial for TAM differentiation and function. Here, we investigate the role of HES1, a downstream target of Notch signaling, in TAM-mediated immunosuppression and explore its potential as a target for cancer immunotherapy. METHODS In this work, we constructed conditional Hes1 knockout mice to selectively delete Hes1 in TAMs. We further analyzed the TME composition, T cell infiltration and activation, and anti-tumor effects in these mice, both alone and in combination with PD-1 checkpoint blockade. RESULTS Our study showed that expression levels of Notch target Hes1 were increase in TAMs and mice with conditional knockout of Hes1 gene in TAMs exhibited decreased tumor growth, with increased infiltration and activation of cytotoxic T cells in tumors. Expression of tumor promoting factors was critically altered in Hes1-conditional KO TAMs, leading to the improved tumor microenvironment. Notably, arginase-1 expression was decreased in Hes1-conditional KO mice. Arg1 is known to deplete arginine and deactivate T cells in the TME. Administration of anti-PD-1 monoclonal antibody inhibited tumor growth to a greater extent in Hes1-conditional KO mice than in WT mice. CONCLUSIONS We identified a pivotal role for the Notch signaling pathway in shaping TAM function, suggesting that T-cell dysfunction in the TME is caused when the Notch target, HES1, in TAMs is upregulated by tumor-associated factors (TAFs), which, in turn, increases the expression of arginase-1. Targeting HES1 in TAMs appears to be a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Myung Sup Kim
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyeokgu Kang
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jung-Hwan Baek
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Moon-Gyu Cho
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Seok-Jun Kim
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
- Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Affiliate Faculty, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
13
|
Félix J, Díaz-Del Cerro E, Baca A, López-Ballesteros A, Gómez-Sánchez MJ, De la Fuente M. Human Supplementation with AM3, Spermidine, and Hesperidin Enhances Immune Function, Decreases Biological Age, and Improves Oxidative-Inflammatory State: A Randomized Controlled Trial. Antioxidants (Basel) 2024; 13:1391. [PMID: 39594533 PMCID: PMC11591374 DOI: 10.3390/antiox13111391] [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: 10/16/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The positive effect of AM3, spermidine, and hesperidin, which have antioxidant and anti-inflammatory properties, on immunity is known, but their effect on the rate of aging, known as biological age (BA), is unclear. This work aims to test if the intake of a blend of AM3 (150 mg), spermidine (0.6 mg), and hesperidin (50 mg) for 2 months could decrease BA and improve immunity, redox, and inflammatory states. For this, 41 participants (30-63 years) were randomly divided into placebo and supplement groups. The supplement group took two capsules daily with AM3, spermidine, and hesperidin for two months, while the placebo group took capsules containing only calcium phosphate and talcum powder. Before and after the treatment, peripheral blood was collected. Immune function was assessed in leukocytes, redox state in whole-blood cells, erythrocytes, and plasma, and cytokine concentration in both mononuclear cell cultures and plasma. Finally, the Immunity Clock model was applied to determine BA. The results show that the intake of this blend improves the immune functions that constitute the Immunity Clock, decreasing BA by 11 years and reducing the oxidative-inflammatory state of the participants. Therefore, this supplement can be proposed as a strategy to rejuvenate BA and achieve healthy aging.
Collapse
Affiliation(s)
- Judith Félix
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (E.D.-D.C.); (A.B.)
- Institute of Investigation Hospital 12 Octubre (Imas12), 28041 Madrid, Spain
| | - Estefanía Díaz-Del Cerro
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (E.D.-D.C.); (A.B.)
- Institute of Investigation Hospital 12 Octubre (Imas12), 28041 Madrid, Spain
| | - Adriana Baca
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (E.D.-D.C.); (A.B.)
| | | | | | - Mónica De la Fuente
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (E.D.-D.C.); (A.B.)
- Institute of Investigation Hospital 12 Octubre (Imas12), 28041 Madrid, Spain
| |
Collapse
|
14
|
Phillips EA, Alharithi YJ, Kadam L, Coussens LM, Kumar S, Maloyan A. Metabolic abnormalities in the bone marrow cells of young offspring born to mothers with obesity. Int J Obes (Lond) 2024; 48:1542-1551. [PMID: 38937647 DOI: 10.1038/s41366-024-01563-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/OBJECTIVES Intrauterine metabolic reprogramming occurs in mothers with obesity during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans born to women with obesity. METHODS Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art flow cytometry, and targeted metabolomics approach coupled with a Seahorse metabolic analyzer. RESULTS We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation. We also show a reduction in levels of amino acids, a phenomenon previously linked to bone marrow aging. Using flow cytometry, we found changes in the immune complexity of bone marrow cells and identified a unique B cell population expressing CD19 and CD11b in the bone marrow of three-week-old offspring of high-fat diet-fed mothers. Our data also revealed increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11bhi B cells. CONCLUSIONS Altogether, we demonstrate that the offspring of mothers with obesity show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
Affiliation(s)
- Elysse A Phillips
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
- The University of California San Francisco, San Francisco, CA, USA
| | - Yem J Alharithi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Leena Kadam
- Department of OB/GYN, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Lisa M Coussens
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sushil Kumar
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| | - Alina Maloyan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
- The University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
15
|
Yoon C, Kim HK, Ham YS, Gil WJ, Mun SJ, Cho E, Yuk JM, Yang CS. Toxoplasma gondii macrophage migration inhibitory factor shows anti- Mycobacterium tuberculosis potential via AZIN1/STAT1 interaction. SCIENCE ADVANCES 2024; 10:eadq0101. [PMID: 39453997 PMCID: PMC11506136 DOI: 10.1126/sciadv.adq0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024]
Abstract
Mycobacterium tuberculosis (MTB) is a pathogenic bacterium, belonging to the family Mycobacteriaceae, that causes tuberculosis (TB). Toxoplasma gondii macrophage migration inhibitory factor (TgMIF), a protein homolog of macrophage migration inhibitory factor, has been explored for its potential to modulate immune responses during MTB infections. We observed that TgMIF that interacts with CD74, antizyme inhibitor 1 (AZIN1), and signal transducer and activator of transcription 1 (STAT1) modulates endocytosis, restoration of mitochondrial function, and macrophage polarization, respectively. These interactions promote therapeutic efficacy in mice infected with MTB, thereby presenting a potential route to host-directed therapy development. Furthermore, TgMIF, in combination with first-line TB drugs, significantly inhibited drug-resistant MTB strains, including multidrug-resistant TB. These results demonstrate that TgMIF is potentially a multifaceted therapeutic agent against TB, acting through immune modulation, enhancement of mitochondrial function, and dependent on STAT1 and AZIN1 pathways.
Collapse
Affiliation(s)
- Chanjin Yoon
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588, South Korea
| | - Hyo Keun Kim
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Yu Seong Ham
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Woo Jin Gil
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Seok-Jun Mun
- Department of Bionano Engineering, Hanyang University, Seoul 04673, South Korea
| | - Euni Cho
- Department of Bionano Engineering, Hanyang University, Seoul 04673, South Korea
| | - Jae-Min Yuk
- Department of Infection Biology and Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
- Department of Medicinal and Life Science, Hanyang University, Ansan 15588, South Korea
| |
Collapse
|
16
|
Zhang X, Guo H, Li X, Tao W, Ma X, Zhang Y, Xiao W. Single-cell omics and machine learning integration to develop a polyamine metabolism-based risk score model in breast cancer patients. J Cancer Res Clin Oncol 2024; 150:473. [PMID: 39441216 PMCID: PMC11499360 DOI: 10.1007/s00432-024-06001-z] [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: 08/30/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Breast cancer remains the leading malignant neoplasm among women globally, posing significant challenges in terms of treatment and prognostic evaluation. The metabolic pathway of polyamines is crucial in breast cancer progression, with a strong association to the increased capabilities of tumor cells for proliferation, invasion, and metastasis. METHODS We used a multi-omics approach combining bulk RNA sequencing and single-cell RNA sequencing (scRNA-seq) to study polyamine metabolism. Data from The Cancer Genome Atlas, Gene Expression Omnibus, and Genotype-Tissue Expression identified 286 differentially expressed genes linked to polyamine pathways in breast cancer. These genes were analyzed using univariate COX and machine learning algorithms to develop a prognostic scoring algorithm. Single-cell RNA sequencing validated the model by examining gene expression heterogeneity at the cellular level. RESULTS Our single-cell analyses revealed distinct subpopulations with different expressions of genes related to polyamine metabolism, highlighting the heterogeneity of the tumor microenvironment. The SuperPC model (a constructed risk score) demonstrated high accuracy when predicting patient outcomes. The immune profiling and functional enrichment analyses revealed that the genes identified play a crucial role in cell cycle control and immune modulation. Single-cell validation confirmed that polyamine metabolism genes were present in specific cell clusters. This highlights their potential as therapeutic targets. CONCLUSIONS This study integrates single cell omics with machine-learning to develop a robust scoring model for breast cancer based on polyamine metabolic pathways. Our findings offer new insights into tumor heterogeneity, and a novel framework to personalize prognosis. Single-cell technologies are being used in this context to enhance our understanding of the complex molecular terrain of breast cancer and support more effective clinical management.
Collapse
Affiliation(s)
- Xiliang Zhang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Hanjie Guo
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xiaolong Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Wei Tao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Xiaoqing Ma
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Yuxing Zhang
- Department of General Surgery, The Sixth Medical Center of PLA General Hospital, 6 Fucheng Road, Haidian, Beijing, 100048, P.R. China.
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| |
Collapse
|
17
|
Ji G, Liu J, Zhao Z, Lan J, Yang Y, Wang Z, Feng H, Ji K, Jiang X, Xia H, Wei G, Zhang Y, Zhang Y, Du X, Wang Y, Yang Y, Liu Z, Zhang K, Mei Q, Sun R, Lu H. Polyamine Anabolism Promotes Chemotherapy-Induced Breast Cancer Stem Cell Enrichment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404853. [PMID: 39058337 PMCID: PMC11516096 DOI: 10.1002/advs.202404853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/27/2024] [Indexed: 07/28/2024]
Abstract
Breast cancer patients may initially benefit from cytotoxic chemotherapy but experience treatment resistance and relapse. Chemoresistant breast cancer stem cells (BCSCs) play a pivotal role in cancer recurrence and metastasis, however, identification and eradication of BCSC population in patients are challenging. Here, an mRNA-based BCSC signature is developed using machine learning strategy to evaluate cancer stemness in primary breast cancer patient samples. Using the BCSC signature, a critical role of polyamine anabolism in the regulation of chemotherapy-induced BCSC enrichment, is elucidated. Mechanistically, two key polyamine anabolic enzymes, ODC1 and SRM, are directly activated by transcription factor HIF-1 in response to chemotherapy. Genetic inhibition of HIF-1-controlled polyamine anabolism blocks chemotherapy-induced BCSC enrichment in vitro and in xenograft mice. A novel specific HIF-1 inhibitor britannin is identified through a natural compound library screening, and demonstrate that coadministration of britannin efficiently inhibits chemotherapy-induced HIF-1 transcriptional activity, ODC1 and SRM expression, polyamine levels, and BCSC enrichment in vitro and in xenograft and autochthonous mouse models. The findings demonstrate the key role of polyamine anabolism in BCSC regulation and provide a new strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Guangyu Ji
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
- School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinan250012China
| | - Jia Liu
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
| | - Zhiqun Zhao
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
| | - Jie Lan
- Department of Radiation OncologyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - You Yang
- Department of Pediatrics (Children Hematological Oncology)Birth Defects and Childhood Hematological Oncology LaboratoryThe Affiliated Hospital of Southwest Medical UniversitySichuan Clinical Research Center for Birth DefectsLuzhou646000China
| | - Zheng Wang
- Department of UrologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan250021China
| | - Huijing Feng
- Cancer Center, Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuan030032China
| | - Kai Ji
- Shandong Helix Matrix Data TechnologyJinan250014China
| | - Xiaofeng Jiang
- Youth League CommitteeQilu HospitalShandong UniversityJinan250012China
| | - Huize Xia
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
| | - Guangyao Wei
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
| | - Yajing Zhang
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
| | - Yuhong Zhang
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
| | - Xinlong Du
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
| | - Yawen Wang
- Department of Breast Surgery, General SurgeryQilu HospitalShandong UniversityJinan250012China
| | - Yuanyuan Yang
- Shandong Artificial Intelligence InstituteQilu University of Technology (Shandong Academy of Sciences)Jinan250399China
| | - Zhaojian Liu
- School of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinan250012China
| | - Kai Zhang
- Department of Breast Surgery, General SurgeryQilu HospitalShandong UniversityJinan250012China
| | - Qi Mei
- Cancer Center, Shanxi Bethune HospitalShanxi Academy of Medical SciencesTongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuan030032China
- Department of Oncology, Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
| | - Rong Sun
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
| | - Haiquan Lu
- The Second Hospital and Advanced Medical Research InstituteCheeloo College of MedicineShandong UniversityJinan250012China
- Key Laboratory for Experimental Teratology of the Ministry of EducationCheeloo College of MedicineShandong UniversityJinan250012China
- Center for Reproductive MedicineShandong UniversityJinan250001China
| |
Collapse
|
18
|
Yadav S, Sapra L, Srivastava RK. Polysaccharides to postbiotics: Nurturing bone health via modulating "gut-immune axis". Int J Biol Macromol 2024; 278:134655. [PMID: 39128750 DOI: 10.1016/j.ijbiomac.2024.134655] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The increasing prevalence of individuals affected by bone pathologies globally has sparked catastrophic concerns. Ankylosing spondylitis, osteoporosis, rheumatoid arthritis, osteoarthritis, and fractures alone impact an estimated 1.71 billion people worldwide. The gut microbiota plays a crucial role in interacting with the host through the synthesis of a diverse range of metabolites called gut-associated metabolites (GAMs), which originate from external dietary substrates or endogenous host compounds. Many metabolic disorders have been linked to alterations in the gut microbiota's activity and composition. The development of metabolic illnesses has been linked to certain microbiota-derived metabolites, such as branched-chain amino acids, bile acids, short-chain fatty acids, tryptophan, trimethylamine N-oxide, and indole derivatives. Moreover, the modulation of gut microbiota through biotics (prebiotics, probiotics and postbiotics) presents a promising avenue for therapeutic intervention. Biotics selectively promote the growth of beneficial gut bacteria, thereby enhancing the production of GAMs with potential beneficial effects on bone metabolism. Understanding the intricate interplay between GAMs, and bone-associated genes through molecular informatics holds significant promise for early diagnosis, prognosis, and novel treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Sumedha Yadav
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
19
|
Chang X, Zhang S, Li C, Zhang H, Yang W, Zhang W, Ye Z, Liang Y, Qiu X, Zeng J. Inhibitory Effect of Lactobacillus Paracasei CMU-Pb-L5 In a Subcutaneous Transplanted Tumor Model of Colorectal Cancer. Int J Med Sci 2024; 21:2525-2536. [PMID: 39439459 PMCID: PMC11492875 DOI: 10.7150/ijms.99646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Lactobacillus paracasei (L.p) is a prevalent probiotic strain within the Lactobacillus genus, which has robust intestinal colonization capabilities. Previous studies have demonstrated the anticancer properties of L.p both in vivo and in vitro. However, the mechanisms underlying its anticancer activity in vivo remain unclear. This study established a subcutaneous transplanted tumor model of colorectal cancer (CRC) in mice to investigate the impact of L.p CMU-Pb-L5. Various parameters including tumor volume, tumor weight, histological alterations in tumor tissue, levels of polyamines and immune-related cytokines in serum, as well as the expression of polyamine metabolism-related and apoptosis-related proteins were evaluated. The results suggested that L.p CMU-Pb-L5 exhibited inhibitory effects on tumor cell proliferation, promotion of tumor cell apoptosis, reduction in polyamine levels, and enhancement of the immune response in CRC mice. To sum up, these results suggested that L.p CMU-Pb-L5 holds promise for potential clinical applications in the treatment of CRC.
Collapse
Affiliation(s)
- Xiaodan Chang
- Department of Neonatology, The Second Central Hospital of Baoding, Baoding 071051, China
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| | - Shaobing Zhang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| | - Cong Li
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
- Dongguan Key Laboratory of Metabolic Immunology and Oral Disease, Department of Stomatology, Dongguan Maternal and Child Health Care Hospital, Dongguan 523000, China
| | - Hailiang Zhang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| | - Weiqing Yang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| | - Weijian Zhang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
- Community health service center of Dongguan Dalang Town, Dongguan 523000, China
| | - Ziyu Ye
- Xinghai Institute of Cell, Guangdong Xianhua Institute for Medical Research, Dongguan 523808, China
| | - Yanfang Liang
- Dongguan Key Laboratory of Molecular Immunopathology, Department of Pathology, Binhaiwan Central Hospital of Dongguan, Dongguan 523000, China
| | - Xianxiu Qiu
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| | - Jincheng Zeng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
- Xinghai Institute of Cell, Guangdong Xianhua Institute for Medical Research, Dongguan 523808, China
| |
Collapse
|
20
|
Pochini L. Involvement of mammalian SoLute Carriers (SLC) in the traffic of polyamines. Front Mol Biosci 2024; 11:1452184. [PMID: 39130372 PMCID: PMC11310933 DOI: 10.3389/fmolb.2024.1452184] [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: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Polyamines interact with different molecular targets to regulate a vast range of cellular processes. A network of enzymes and transport systems is crucial for the maintenance of polyamine homeostasis. Indeed, polyamines after synthesis must be distributed to the various tissues and some intracellular organelles. Differently from the well characterized enzymes devoted to polyamine synthesis, the transport systems are not unequivocally identified or characterized. Besides some ATPases which have been identified as polyamine transporters, much less is known about solute carriers (SLC) involved in the transport of these compounds. Only two SLCs have been unequivocally identified as polyamine transporters: SLC18B1 (VPAT) and SLC22A4 (OCTN1). Transport studies have been performed with cells transfected with the cDNAs encoding the two and other SLCs or, in the case of OCTN1, also by in vitro assay using proteoliposomes harboring the recombinant human protein. According to the role proposed for OCTN1, polyamines have been associated with prolonged and quality of life. This review provides an update on the most recent findings concerning the polyamine transporters or the prediction of the putative ones.
Collapse
Affiliation(s)
- Lorena Pochini
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze Della Terra), University of Calabria, Rende, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Bari, Italy
| |
Collapse
|
21
|
Cruciani S, Coradduzza D, Balzano F, Garroni G, Azara E, Pala R, Delitala AP, Madonia M, Tedde A, Capobianco G, Petrillo M, Angelucci C, Carru C, Ventura C, Maioli M. Modulation of adipose-derived stem cell behavior by prostate pathology-associated plasma: insights from in vitro exposure. Sci Rep 2024; 14:14765. [PMID: 38926454 PMCID: PMC11208502 DOI: 10.1038/s41598-024-64625-0] [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: 03/16/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are promising in regenerative medicine. Their proliferation, survival and activation are influenced by specific signals within their microenvironment, also known as niche. The stem cell niche is regulated by complex interactions between multiple cell types. When transplanted in a specific area, ADSCs can secrete several immunomodulatory factors. At the same time, a tumor microenvironment can influence stem cell behavior, modulating proliferation and their ability to differentiate into a specific phenotype. Whitin this context, we exposed ADSCs to plasma samples derived from human patients diagnosed with prostate cancer (PC), or precancerous lesions (PL), or benign prostatic hyperplasia (BPH) for 4, 7 or 10 days. We then analyzed the expression of main stemness-related markers and cell-cycle regulators. We also measured cytokine production and polyamine secretion in culture medium and evaluated cell morphology and collagen production by confocal microscopy. The results obtained from this study show significant changes in the morphology of ADSCs exposed to plasma samples, especially in the presence of prostate cancer plasma, suggesting important implications in the use of ADSCs for the development of new treatments and application in regenerative medicine.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Donatella Coradduzza
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Emanuela Azara
- Institute of Biomolecular Chemistry, National Research Council, 07100, Sassari, Italy
| | - Renzo Pala
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Alessandro P Delitala
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Massimo Madonia
- Department of Clinical and Experimental Medicine, Urologic Clinic, University of Sassari, Sassari, Italy
| | - Alessandro Tedde
- Department of Clinical and Experimental Medicine, Urologic Clinic, University of Sassari, Sassari, Italy
| | - Giampiero Capobianco
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Marco Petrillo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Cecilia Angelucci
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
- Medical Oncology Unit, University Hospital (AOU) of Sassari, 07100, Sassari, Italy
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, Istituto Nazionale Biostrutture E Biosistemi (INBB)-Eldor Lab, Via Corticella 183, 40128, Bologna, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy.
- Center for Developmental Biology and Reprogramming-CEDEBIOR, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy.
| |
Collapse
|
22
|
Holbert CE, Casero RA, Stewart TM. Polyamines: the pivotal amines in influencing the tumor microenvironment. Discov Oncol 2024; 15:173. [PMID: 38761252 PMCID: PMC11102423 DOI: 10.1007/s12672-024-01034-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 05/11/2024] [Indexed: 05/20/2024] Open
Abstract
Cellular proliferation, function and survival is reliant upon maintaining appropriate intracellular polyamine levels. Due to increased metabolic needs, cancer cells elevate their polyamine pools through coordinated metabolism and uptake. High levels of polyamines have been linked to more immunosuppressive tumor microenvironments (TME) as polyamines support the growth and function of many immunosuppressive cell types such as MDSCs, macrophages and regulatory T-cells. As cancer cells and other pro-tumorigenic cell types are highly dependent on polyamines for survival, pharmacological modulation of polyamine metabolism is a promising cancer therapeutic strategy. This review covers the roles of polyamines in various cell types of the TME including both immune and stromal cells, as well as how competition for nutrients, namely polyamine precursors, influences the cellular landscape of the TME. It also details the use of polyamines as biomarkers and the ways in which polyamine depletion can increase the immunogenicity of the TME and reprogram tumors to become more responsive to immunotherapy.
Collapse
Affiliation(s)
- Cassandra E Holbert
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert A Casero
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tracy Murray Stewart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
Murthy D, Attri KS, Suresh V, Rajacharya GH, Valenzuela CA, Thakur R, Zhao J, Shukla SK, Chaika NV, LaBreck D, Rao CV, Hollingsworth MA, Mehla K, Singh PK. The MUC1-HIF-1α signaling axis regulates pancreatic cancer pathogenesis through polyamine metabolism remodeling. Proc Natl Acad Sci U S A 2024; 121:e2315509121. [PMID: 38547055 PMCID: PMC10998584 DOI: 10.1073/pnas.2315509121] [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: 09/06/2023] [Accepted: 02/22/2024] [Indexed: 04/02/2024] Open
Abstract
Dysregulation of polyamine metabolism has been implicated in cancer initiation and progression; however, the mechanism of polyamine dysregulation in cancer is not fully understood. In this study, we investigated the role of MUC1, a mucin protein overexpressed in pancreatic cancer, in regulating polyamine metabolism. Utilizing pancreatic cancer patient data, we noted a positive correlation between MUC1 expression and the expression of key polyamine metabolism pathway genes. Functional studies revealed that knockdown of spermidine/spermine N1-acetyltransferase 1 (SAT1), a key enzyme involved in polyamine catabolism, attenuated the oncogenic functions of MUC1, including cell survival and proliferation. We further identified a regulatory axis whereby MUC1 stabilized hypoxia-inducible factor (HIF-1α), leading to increased SAT1 expression, which in turn induced carbon flux into the tricarboxylic acid cycle. MUC1-mediated stabilization of HIF-1α enhanced the promoter occupancy of the latter on SAT1 promoter and corresponding transcriptional activation of SAT1, which could be abrogated by pharmacological inhibition of HIF-1α or CRISPR/Cas9-mediated knockout of HIF1A. MUC1 knockdown caused a significant reduction in the levels of SAT1-generated metabolites, N1-acetylspermidine and N8-acetylspermidine. Given the known role of MUC1 in therapy resistance, we also investigated whether inhibiting SAT1 would enhance the efficacy of FOLFIRINOX chemotherapy. By utilizing organoid and orthotopic pancreatic cancer mouse models, we observed that targeting SAT1 with pentamidine improved the efficacy of FOLFIRINOX, suggesting that the combination may represent a promising therapeutic strategy against pancreatic cancer. This study provides insights into the interplay between MUC1 and polyamine metabolism, offering potential avenues for the development of treatments against pancreatic cancer.
Collapse
Affiliation(s)
- Divya Murthy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
| | - Kuldeep S. Attri
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
| | - Voddu Suresh
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Girish H. Rajacharya
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Carlos A. Valenzuela
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Ravi Thakur
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Junzhang Zhao
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Surendra K. Shukla
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Nina V. Chaika
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
| | - Drew LaBreck
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Chinthalapally V. Rao
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
| | - Kamiya Mehla
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| | - Pankaj K. Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE68198-5950
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK73104
| |
Collapse
|
24
|
Qiao J, Cai W, Wang K, Haubruge E, Dong J, El-Seedi HR, Xu X, Zhang H. New Insights into Identification, Distribution, and Health Benefits of Polyamines and Their Derivatives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5089-5106. [PMID: 38416110 DOI: 10.1021/acs.jafc.3c08556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Polyamines and their derivatives are ubiquitously present in free or conjugated forms in various foods from animal, plant, and microbial origins. The current knowledge of free polyamines in foods and their contents is readily available; furthermore, conjugated polyamines generate considerable recent research interest due to their potential health benefits. The structural diversity of conjugated polyamines results in challenging their qualitative and quantitative analysis in food. Herein, we review and summarize the knowledge published on polyamines and their derivatives in foods, including their identification, sources, quantities, and health benefits. Particularly, facing the inherent challenges of isomer identification in conjugated polyamines, this paper provides a comprehensive overview of conjugated polyamines' structural characteristics, including the cleavage patterns and characteristic ion fragments of MS/MS for isomer identification. Free polyamines are present in all types of food, while conjugated polyamines are limited to plant-derived foods. Spermidine is renowned for antiaging properties, acclaimed as antiaging vitamins. Conjugated polyamines highlight their anti-inflammatory properties and have emerged as the mainstream drugs for antiprostatitis. This paper will likely help us gain better insight into polyamines and their derivatives to further develop functional foods and personalized nutraceuticals.
Collapse
Affiliation(s)
- Jiangtao Qiao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
- Terra Research Center, Gembloux Agro-Bio Tech, University of Liege, Gembloux 5030, Belgium
| | - Wenwen Cai
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
- College of Food Engineering, Harbin University of Commerce, Harbin 155023, China
| | - Kai Wang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
- Terra Research Center, Gembloux Agro-Bio Tech, University of Liege, Gembloux 5030, Belgium
| | - Eric Haubruge
- Terra Research Center, Gembloux Agro-Bio Tech, University of Liege, Gembloux 5030, Belgium
| | - Jie Dong
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
- Key Laboratory of Bee Products for Quality and Safety Control, Ministry of Agriculture and Rural Affairs, Beijing 100093, China
| | - Hesham R El-Seedi
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, BMC, Uppsala University, Box 591, SE 75124 Uppsala, Sweden
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia
| | - Xiang Xu
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Hongcheng Zhang
- Key Laboratory of Bee Products for Quality and Safety Control, Ministry of Agriculture and Rural Affairs, Beijing 100093, China
| |
Collapse
|
25
|
Alina M, Phillips E, Alharithi Y, Kadam L, Coussens L, Kumar S. Metabolic abnormalities in the bone marrow cells of young offspring born to obese mothers. RESEARCH SQUARE 2024:rs.3.rs-3830161. [PMID: 38313293 PMCID: PMC10836107 DOI: 10.21203/rs.3.rs-3830161/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Intrauterine metabolic reprogramming occurs in obese mothers during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans born to obese women. Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art targeted metabolomics approach coupled with a Seahorse metabolic analyzer. We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation. We also found a reduction in levels of amino acids, a phenomenon previously linked to bone marrow aging. Using flow cytometry, we identified a unique B cell population expressing CD19 and CD11b in the bone marrow of three-week-old offspring of high-fat diet-fed mothers, and found increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11bhi B cells. Altogether, we demonstrate that the offspring of obese mothers show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
|
26
|
Al-Qahtani AA, Alhamlan FS, Al-Qahtani AA. Pro-Inflammatory and Anti-Inflammatory Interleukins in Infectious Diseases: A Comprehensive Review. Trop Med Infect Dis 2024; 9:13. [PMID: 38251210 PMCID: PMC10818686 DOI: 10.3390/tropicalmed9010013] [Citation(s) in RCA: 73] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/29/2023] [Accepted: 12/10/2023] [Indexed: 01/23/2024] Open
Abstract
Interleukins (ILs) are signaling molecules that are crucial in regulating immune responses during infectious diseases. Pro-inflammatory ILs contribute to the activation and recruitment of immune cells, whereas anti-inflammatory ILs help to suppress excessive inflammation and promote tissue repair. Here, we provide a comprehensive overview of the role of pro-inflammatory and anti-inflammatory ILs in infectious diseases, with a focus on the mechanisms underlying their effects, their diagnostic and therapeutic potential, and emerging trends in IL-based therapies.
Collapse
Affiliation(s)
- Arwa A. Al-Qahtani
- Department of Family Medicine, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Fatimah S. Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Ahmed Ali Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| |
Collapse
|
27
|
Ridnour LA, Cheng RYS, Heinz WF, Pore M, Gonzalez AL, Femino EL, Moffat R, Wink AL, Imtiaz F, Coutinho L, Butcher D, Edmondson EF, Rangel MC, Wong STC, Lipkowitz S, Glynn S, Vitek MP, McVicar DW, Li X, Anderson SK, Paolocci N, Hewitt SM, Ambs S, Billiar TR, Chang JC, Lockett SJ, Wink DA. Spatial analysis of NOS2 and COX2 interaction with T-effector cells reveals immunosuppressive landscapes associated with poor outcome in ER- breast cancer patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572867. [PMID: 38187660 PMCID: PMC10769421 DOI: 10.1101/2023.12.21.572867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Multiple immunosuppressive mechanisms exist in the tumor microenvironment that drive poor outcomes and decrease treatment efficacy. The co-expression of NOS2 and COX2 is a strong predictor of poor prognosis in ER- breast cancer and other malignancies. Together, they generate pro-oncogenic signals that drive metastasis, drug resistance, cancer stemness, and immune suppression. Using an ER- breast cancer patient cohort, we found that the spatial expression patterns of NOS2 and COX2 with CD3+CD8+PD1- T effector (Teff) cells formed a tumor immune landscape that correlated with poor outcome. NOS2 was primarily associated with the tumor-immune interface, whereas COX2 was associated with immune desert regions of the tumor lacking Teff cells. A higher ratio of NOS2 or COX2 to Teff was highly correlated with poor outcomes. Spatial analysis revealed that regional clustering of NOS2 and COX2 was associated with stromal-restricted Teff, while only COX2 was predominant in immune deserts. Examination of other immunosuppressive elements, such as PDL1/PD1, Treg, B7H4, and IDO1, revealed that PDL1/PD1, Treg, and IDO1 were primarily associated with restricted Teff, whereas B7H4 and COX2 were found in tumor immune deserts. Regardless of the survival outcome, other leukocytes, such as CD4 T cells and macrophages, were primarily in stromal lymphoid aggregates. Finally, in a 4T1 model, COX2 inhibition led to a massive cell infiltration, thus validating the hypothesis that COX2 is an essential component of the Teff exclusion process and, thus, tumor evasion. Our study indicates that NOS2/COX2 expression plays a central role in tumor immunosuppression. Our findings indicate that new strategies combining clinically available NOS2/COX2 inhibitors with various forms of immune therapy may open a new avenue for the treatment of aggressive ER-breast cancers.
Collapse
Affiliation(s)
- Lisa A Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Robert Y S Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - William F Heinz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Milind Pore
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research
| | - Ana L Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Elise L Femino
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Rebecca Moffat
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Adelaide L Wink
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Fatima Imtiaz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Leandro Coutinho
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Donna Butcher
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
| | - Elijah F Edmondson
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
| | - M Cristina Rangel
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Sharon Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | | | - Daniel W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Stephen K Anderson
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Basic Science Program, Frederick National Laboratory for Cancer Research
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University, and Department of Biomedical Sciences, University of Padova, Italy
- Laboratory of Pathology CCR, NCI, NIH
| | | | - Stefan Ambs
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Timothy R Billiar
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX
| | - Jenny C Chang
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
- Houston Methodist Weill Cornell Medical College, Houston TX
- Women's Malignancies Branch, CCR, NCI, NIH
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
- (Mike Duke)
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
- Basic Science Program, Frederick National Laboratory for Cancer Research
- Division of Cardiology, Department of Medicine, Johns Hopkins University, and Department of Biomedical Sciences, University of Padova, Italy
- Laboratory of Pathology CCR, NCI, NIH
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - David A Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| |
Collapse
|
28
|
Phillips E, Alharithi Y, Kadam L, Coussens LM, Kumar S, Maloyan A. Metabolic abnormalities in the bone marrow cells of young offspring born to obese mothers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569274. [PMID: 38077037 PMCID: PMC10705475 DOI: 10.1101/2023.11.29.569274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Intrauterine metabolic reprogramming occurs in obese mothers during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans. Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art targeted metabolomics approach coupled with a Seahorse metabolic analyzer. We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation, and reduction in levels of amino acids, a phenomenon previously linked to aging. Furthermore, in the bone marrow of three-week-old offspring of high-fat diet-fed mothers, we identified a unique B cell population expressing CD19 and CD11b, and found increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11b hi B cells, with all the populations being significantly more abundant in offspring of dams fed HFD but not a regular diet. Altogether, we demonstrate that the offspring of obese mothers show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
|
29
|
Shao T, Hsu R, Rafizadeh DL, Wang L, Bowlus CL, Kumar N, Mishra J, Timilsina S, Ridgway WM, Gershwin ME, Ansari AA, Shuai Z, Leung PSC. The gut ecosystem and immune tolerance. J Autoimmun 2023; 141:103114. [PMID: 37748979 DOI: 10.1016/j.jaut.2023.103114] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023]
Abstract
The gastrointestinal tract is home to the largest microbial population in the human body. The gut microbiota plays significant roles in the development of the gut immune system and has a substantial impact on the maintenance of immune tolerance beginning in early life. These microbes interact with the immune system in a dynamic and interdependent manner. They generate immune signals by presenting a vast repertoire of antigenic determinants and microbial metabolites that influence the development, maturation and maintenance of immunological function and homeostasis. At the same time, both the innate and adaptive immune systems are involved in modulating a stable microbial ecosystem between the commensal and pathogenic microorganisms. Hence, the gut microbial population and the host immune system work together to maintain immune homeostasis synergistically. In susceptible hosts, disruption of such a harmonious state can greatly affect human health and lead to various auto-inflammatory and autoimmune disorders. In this review, we discuss our current understanding of the interactions between the gut microbiota and immunity with an emphasis on: a) important players of gut innate and adaptive immunity; b) the contribution of gut microbial metabolites; and c) the effect of disruption of innate and adaptive immunity as well as alteration of gut microbiome on the molecular mechanisms driving autoimmunity in various autoimmune diseases.
Collapse
Affiliation(s)
- Tihong Shao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China; Division of Rheumatology/Allergy and Clinical Immunology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Ronald Hsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Desiree L Rafizadeh
- Division of Rheumatology/Allergy and Clinical Immunology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Li Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Christopher L Bowlus
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Narendra Kumar
- Department of Pharmaceutical Science, ILR-College of Pharmacy, Texas A&M University, 1010 W. Ave B. MSC 131, Kingsville, TX, 78363, USA
| | - Jayshree Mishra
- Department of Pharmaceutical Science, ILR-College of Pharmacy, Texas A&M University, 1010 W. Ave B. MSC 131, Kingsville, TX, 78363, USA
| | - Suraj Timilsina
- Division of Rheumatology/Allergy and Clinical Immunology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - William M Ridgway
- Division of Rheumatology/Allergy and Clinical Immunology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - M Eric Gershwin
- Division of Rheumatology/Allergy and Clinical Immunology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Aftab A Ansari
- Division of Rheumatology/Allergy and Clinical Immunology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Zongwen Shuai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Patrick S C Leung
- Division of Rheumatology/Allergy and Clinical Immunology, Department of Internal Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
30
|
Zhang Y, Zhao X, Qin Y, Li X, Chang Y, Shi Z, Song M, Sun W, Xiao J, Li Z, Qing G. Order-order assembly transition-driven polyamines detection based on iron-sulfur complexes. Commun Chem 2023; 6:146. [PMID: 37420027 PMCID: PMC10328931 DOI: 10.1038/s42004-023-00942-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023] Open
Abstract
Innovative modes of response can greatly push forward chemical sensing processes and subsequently improve sensing performance. Classical chemical sensing modes seldom involve the transition of a delicate molecular assembly during the response. Here, we display a sensing mode for polyamine detection based on an order-order transition of iron-sulfur complexes upon their assembly. Strong validation proves that the unique order-order transition of the assemblies is the driving force of the response, in which the polyamine captures the metal ion of the iron-sulfur complex, leading it to decompose into a metal-polyamine product, accompanied by an order-order transition of the assemblies. This mechanism makes the detection process more intuitive and selective, and remarkably improves the detection efficiency, achieving excellent polyamines specificity, second-level response, convenient visual detection, and good recyclability of the sensing system. Furthermore, this paper also provides opportunities for the further application of the iron-sulfur platform in environment-related fields.
Collapse
Affiliation(s)
- Yahui Zhang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xiangyu Zhao
- Sixth Laboratory, Sinopec Dalian (Fushun) Research Institute of Petroleum and Petrochemicals, 96 Nankai Road, Dalian, 116045, P. R. China
| | - Yue Qin
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xiaopei Li
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Yongxin Chang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Zhenqiang Shi
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Mengyuan Song
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Wenjing Sun
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Jie Xiao
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Zan Li
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Guangyan Qing
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China.
- College of Chemistry and Chemical Engineering, Wuhan Textile University, 1 Sunshine Road, Wuhan, 430200, P. R. China.
| |
Collapse
|
31
|
Hansakon A, Jeerawattanawart S, Angkasekwinai P. Differential and cooperative effects of IL-25 and IL-33 on T helper cells contribute to cryptococcal virulence and brain infection. Sci Rep 2023; 13:9895. [PMID: 37337050 DOI: 10.1038/s41598-023-37158-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023] Open
Abstract
The epithelial cell-derived cytokines IL-33 and IL-25 are important mediators in driving type-2 inflammation during C. neoformans infection. Nevertheless, the impact of these cytokines in regulating host T helper cell response during C. neoformans infection is still unclear. We observed that C. neoformans infection promoted a predominant increase of T helper cells that co-expressed IL-25 and IL-33 receptors within the lung during the late infection phase. A comparative transcriptomic analysis of effector T helper cells co-treated with IL-25 and IL-33 revealed a cooperative effect of these cytokines in promoting IL-13 gene expression. Without IL-25 receptor signaling, IL-33 treatment upregulated Th1-associated genes and genes associated with nucleotide metabolism. By contrast, IL-25 had a unique effect in enhancing type-2 cytokines IL-5 and IL-9 and chemokine CCL24, as well as genes in the pathways that are associated with L-arginine metabolisms. Interestingly, this pathogenic T helper cell population that expressed IL-25 and IL-33 receptors was greatly enriched in mice that were infected with high cryptococcal virulence and associated with fungal burdens in the brain. Therefore, our data further provide the additional function of IL-25 and IL-33 in potentiating cryptococcal brain dissemination.
Collapse
Affiliation(s)
- Adithap Hansakon
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
| | - Siranart Jeerawattanawart
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand
- Graduate Program in Biomedical Science, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, 12120, Thailand.
- Research Unit in Molecular Pathogenesis and Immunology of Infectious Diseases, Thammasat University, Pathum Thani, 12120, Thailand.
| |
Collapse
|
32
|
Wang N, Chai M, Zhu L, Liu J, Yu C, Huang X. Development and validation of polyamines metabolism-associated gene signatures to predict prognosis and immunotherapy response in lung adenocarcinoma. Front Immunol 2023; 14:1070953. [PMID: 37334367 PMCID: PMC10272553 DOI: 10.3389/fimmu.2023.1070953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Background Polyamines metabolism is closely related to tumor development and progression, as well as tumor microenvironment (TME). In this study, we focused on exploring whether polyamines metabolism-associated genes would provide prognosis and immunotherapy response prediction in lung adenocarcinoma (LUAD). Methods The expression profile data of polyamines metabolism-associated genes were acquired from the Cancer Genome Atlas (TCGA) database. Utilizing the least absolute shrinkage and selection operator (LASSO) algorithm, we created a risk score model according to polyamines metabolism-associated gene signatures. Meanwhile, an independent cohort (GSE72094) was employed to validate this model. Through the univariate and multivariate Cox regression analyses, the independent prognostic factors were identified. Subsequently, quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect their expression in LUAD cells. By consensus clustering analysis, polyamines metabolism-associated subgroups were determined in LUAD patients, with differential gene expression, prognosis, and immune characteristics analyses explored. Results A total of 59 polyamines metabolism genes were collected for this study, of which 14 genes were identified for the construction of risk score model using LASSO method. High- and low- risk groups of LUAD patients in TCGA cohort were distinguished via this model, and high-risk group presented dismal clinical outcomes. The same prognostic prediction of this model had been also validated in GSE72094 cohort. Meanwhile, three independent prognostic factors (PSMC6, SMOX, SMS) were determined for constructing the nomogram, and they were all upregulated in LUAD cells. In addition, two distinct subgroups (C1 and C2) were identified in LUAD patients. Comparing the two subgroups, 291 differentially expressed genes (DEGs) were acquired, mainly enriching in organelle fission, nuclear division, and cell cycle. Comparing to C1 subgroup, the patients in C2 subgroup had favorable clinical outcomes, increased immune cells infiltration, and effective immunotherapy response. Conclusion This study identified polyamines metabolism-associated gene signatures for predicting the patients' survival, and they were also linked to immune cells infiltration and immunotherapy response in LUAD patients.
Collapse
Affiliation(s)
- Ning Wang
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Mengyu Chai
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Lingye Zhu
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Jingjing Liu
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Chang Yu
- Intervention Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| |
Collapse
|
33
|
Wetzel TJ, Erfan SC, Figueroa LD, Wheeler LM, Ananieva EA. Crosstalk between arginine, glutamine, and the branched chain amino acid metabolism in the tumor microenvironment. Front Oncol 2023; 13:1186539. [PMID: 37274280 PMCID: PMC10235471 DOI: 10.3389/fonc.2023.1186539] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Arginine, glutamine, and the branched chain amino acids (BCAAs) are a focus of increased interest in the field of oncology due to their importance in the metabolic reprogramming of cancer cells. In the tumor microenvironment (TME), these amino acids serve to support the elevated biosynthetic and energy demands of cancer cells, while simultaneously maintaining the growth, homeostasis, and effector function of tumor-infiltrating immune cells. To escape immune destruction, cancer cells utilize a variety of mechanisms to suppress the cytotoxic activity of effector T cells, facilitating T cell exhaustion. One such mechanism is the ability of cancer cells to overexpress metabolic enzymes specializing in the catabolism of arginine, glutamine, and the BCAAs in the TME. The action of such enzymes supplies cancer cells with metabolic intermediates that feed into the TCA cycle, supporting energy generation, or providing precursors for purine, pyrimidine, and polyamine biosynthesis. Armed with substantial metabolic flexibility, cancer cells redirect amino acids from the TME for their own advantage and growth, while leaving the local infiltrating effector T cells deprived of essential nutrients. This review addresses the metabolic pressure that cancer cells exert over immune cells in the TME by up-regulating amino acid metabolism, while discussing opportunities for targeting amino acid metabolism for therapeutic intervention. Special emphasis is given to the crosstalk between arginine, glutamine, and BCAA metabolism in affording cancer cells with metabolic dominance in the TME.
Collapse
Affiliation(s)
| | | | | | | | - Elitsa A. Ananieva
- Ananieva Laboratory, Biochemistry and Nutrition Department, Des Moines University, Des Moines, IA, United States
| |
Collapse
|
34
|
Zhang Y, Chen R, Zhang D, Qi S, Liu Y. Metabolite interactions between host and microbiota during health and disease: Which feeds the other? Biomed Pharmacother 2023; 160:114295. [PMID: 36709600 DOI: 10.1016/j.biopha.2023.114295] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 01/30/2023] Open
Abstract
Metabolites produced by the host and microbiota play a crucial role in how human bodies develop and remain healthy. Most of these metabolites are produced by microbiota and hosts in the digestive tract. Metabolites in the gut have important roles in energy metabolism, cellular communication, and host immunity, among other physiological activities. Although numerous host metabolites, such as free fatty acids, amino acids, and vitamins, are found in the intestine, metabolites generated by gut microbiota are equally vital for intestinal homeostasis. Furthermore, microbiota in the gut is the sole source of some metabolites, including short-chain fatty acids (SCFAs). Metabolites produced by microbiota, such as neurotransmitters and hormones, may modulate and significantly affect host metabolism. The gut microbiota is becoming recognized as a second endocrine system. A variety of chronic inflammatory disorders have been linked to aberrant host-microbiota interplays, but the precise mechanisms underpinning these disturbances and how they might lead to diseases remain to be fully elucidated. Microbiome-modulated metabolites are promising targets for new drug discovery due to their endocrine function in various complex disorders. In humans, metabolotherapy for the prevention or treatment of various disorders will be possible if we better understand the metabolic preferences of bacteria and the host in specific tissues and organs. Better disease treatments may be possible with the help of novel complementary therapies that target host or bacterial metabolism. The metabolites, their physiological consequences, and functional mechanisms of the host-microbiota interplays will be highlighted, summarized, and discussed in this overview.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anethesiology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - Rui Chen
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - DuoDuo Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin Province 130021, People's Republic of China.
| | - Shuang Qi
- Department of Anethesiology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - Yan Liu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| |
Collapse
|
35
|
Li C, Zhang W, Chang X, Di X, Xie Q, Lin B, Zhang H, Ye Z, Lan M, Lian J, Zhang H, Qiu X, Zeng J, Huang M. The upregulation of peripheral blood polyamine metabolites spermidine and spermine in children with hand, foot, mouth disease is related to enterovirus 71 capsid protein VP1, but not VP4. Transl Pediatr 2023; 12:194-207. [PMID: 36891375 PMCID: PMC9986783 DOI: 10.21037/tp-23-41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/17/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Hand, foot, and mouth disease (HFMD) is a common viral childhood illness caused most commonly by enterovirus 71 (EV71) and coxsackievirus A16. The pathogenesis of EV71 has been extensively studied, and the regulation of the host immune response is suspected to aggravate the serious complications induced by EV71. Our previous research showed that EV71 infection significantly increased the release of circulating interleukin (IL)-6, IL-10, IL-13, and IL-27. Notably, these cytokines are related to the EV71 infection risk and clinical stage. Polyamines are compounds that are ubiquitous in mammalian cells and play a key role in various cellular processes. Several studies have shown that targeting polyamine metabolic pathways can reduce infections caused by viruses. However, the significance of polyamine metabolism in EV71 infection remains largely unknown. METHODS Serum samples from 82 children with HFMD and 70 healthy volunteers (HVs) were collected to determine the polyamine metabolites spermidine (SPD) and spermine (SPM), and IL-6 levels. In addition, peripheral blood mononuclear cells (PBMCs) were treated with EV71 viral protein 1 (VP1) and EV71 VP4, and the cells and supernatant were then collected to analyze the expression of polyamine metabolism-related enzymes by western blot. The data were analyzed using GraphPad Prism 7.0 software (USA). RESULTS The serum polyamine metabolites SPD and SPM were elevated in the HFMD patients, especially in the EV71-infected children. Further, a positive correlation was found between serum SPD and IL-6 levels in the EV71-infected children. We also found that the upregulation of peripheral blood polyamine metabolites in the EV71-infected HFMD children was related to EV71 capsid protein VP1, but not VP4. VP1 may promote the expression of polyamine metabolism-related enzymes and promote the production of polyamine metabolites, thereby upregulating the SPD/nuclear factor kappa B/IL-6 signaling pathway. However, VP4 has the opposite effect in this process. CONCLUSIONS Our results suggest that EV71 capsid protein may regulate the polyamine metabolic pathways of infected cells in a variety of ways. This study provides insights into the mechanism of EV71 infection and polyamine metabolism and has good reference value for the development of EV71 vaccine.
Collapse
Affiliation(s)
- Cong Li
- Department of Stomatology, Dongguan Maternal and Child Health Care Hospital, Dongguan, China
| | - Weijian Zhang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Xiaodan Chang
- Department of Neonatology, The Second Central Hospital of Baoding City, Baoding, China
| | - Xiaohua Di
- Department of Pediatrics, Dongguan People's Hospital, Dongguan, China
| | - Qi Xie
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Bihua Lin
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Hui Zhang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou, China
| | - Ziyu Ye
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China.,Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
| | - Minsheng Lan
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Jiachun Lian
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Hailiang Zhang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Xianxiu Qiu
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Jincheng Zeng
- Department of Stomatology, Dongguan Maternal and Child Health Care Hospital, Dongguan, China.,Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China.,Dongguan Metabolite Analysis Engineering Technology Center of Cells for Medical Use, Guangdong Xinghai Institute of Cell, Dongguan, China
| | - Mingyuan Huang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| |
Collapse
|