1
|
Mathiesen H, Juul-Madsen K, Tramm T, Vorup-Jensen T, Møller HJ, Etzerodt A, Andersen MN. Prognostic value of CD163 + macrophages in solid tumor malignancies: A scoping review. Immunol Lett 2025; 272:106970. [PMID: 39778658 DOI: 10.1016/j.imlet.2025.106970] [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: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Tumor-associated macrophages (TAMs) play crucial roles in development and progression of malignant diseases. Notably, CD163+ TAMs likely perform specific pro-tumorigenic functions, suggesting that this subset may serve as both prognostic biomarkers and targets for future anti-cancer therapy. We conducted a scoping review to map the current knowledge on the prognostic role of CD163+ TAMs in the five most lethal cancers worldwide: Lung, colorectal, gastric, liver, and breast cancer. For all cancer types, most studies showed that high tumoral presence of CD163+ cells was associated with poor patient outcome, and this association was more frequently observed when CD163+ cells were measured at the tumor periphery compared to more central parts of the tumor. These results support that CD163+ TAMs represent a biomarker of poor patient outcome across a variety of solid tumors, and highlight the relevance of further investigations of CD163+ TAMs as targets of future immunotherapies.
Collapse
Affiliation(s)
- Henriette Mathiesen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian Juul-Madsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Trine Tramm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Holger Jon Møller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Morten Nørgaard Andersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Hematology, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
2
|
Moura T, Laranjeira P, Caramelo O, Gil AM, Paiva A. Breast Cancer and Tumor Microenvironment: The Crucial Role of Immune Cells. Curr Oncol 2025; 32:143. [PMID: 40136347 PMCID: PMC11941043 DOI: 10.3390/curroncol32030143] [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: 01/15/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer is the most common type of cancer in women and the second leading cause of death by cancer. Despite recent advances, the mortality rate remains high, underlining the need to develop new therapeutic approaches. The complex interaction between cancer cells and the tumor microenvironment (TME) is crucial in determining tumor progression, therapy response, and patient prognosis. Understanding the role of immune cells in carcinogenesis and tumor progression can help improve targeted therapeutic options, increasing the likelihood of a favorable prognosis. Therefore, this review aims to critically analyze the complex interaction between tumor cells and immune cells, emphasizing the clinical and therapeutic implications. Additionally, we explore advances in immunotherapies, with a focus on immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Tânia Moura
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Group of Environmental Genetics of Oncobiology (CIMAGO), Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Center of Neurosciences and Cell (CNC), University of Coimbra, 3000-504 Coimbra, Portugal
| | - Olga Caramelo
- Gynecology Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-075 Coimbra, Portugal;
| | - Ana M. Gil
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Group of Environmental Genetics of Oncobiology (CIMAGO), Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Ciências Biomédicas Laboratoriais, Instituto Politécnico de Coimbra, ESTESC—Coimbra Health School, 3046-854 Coimbra, Portugal
| |
Collapse
|
3
|
Oner G, Praet MM, Stoop H, Devi GR, Canturk NZ, Altintas S, Van Berckelaer C, Berneman Z, Tjalma W, Koljenovic S, van Dam PA. Tumor Microenvironment Modulation by Tumor-Associated Macrophages: Implications for Neoadjuvant Chemotherapy Response in Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:211-224. [PMID: 40008212 PMCID: PMC11853881 DOI: 10.2147/bctt.s493085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025]
Abstract
Background Tumor-associated macrophages (TAMs) constitute an important part of the tumor microenvironment of breast cancer (BC), and they play an essential role in modulating tumor growth and invasion. However, the role of TAMs in neoadjuvant chemotherapy (NAC) has not been fully elucidated. Therefore, the aim of this study was to assess the function of TAM subtypes and investigate their role in the response to NAC in BC. Methods Presence of TAMs was examined immunohistochemically (IHC) in pre- and post- NAC treatment tumor tissue in a cohort of 138 BC patients. IHC staining with monoclonal antibodies for CD68 and CD163 were performed. Positivity was defined as staining > 1% TAMs in stroma and tumor cell nests. Response to NAC was evaluated according to tumor size change and Residual Cancer Burden (RCB) index. Results CD68+ and CD163+ TAMs decreased significantly in both the stroma and tumor nests (TN) after NAC. The median CD68+ TAMs in the stroma decreased significantly from 5% to 1% (p < 0.005), while CD163+ TAMs showed a marked reduction from 20% to 5% (p < 0.001). Post-NAC, the persistence of CD68+ and CD163+ TAMs in the stroma was strongly correlated with larger residual tumor size (p < 0.005 and p < 0.001, respectively). Changes in CD163+ TAM levels in the stroma were significantly associated with RCB classes (p < 0.005). Pre-NAC, CD163+ TAMs in the stroma and TN showed a significant association with TILs; however, no correlations with TILs were observed post-NAC. Conclusion This study highlights the critical role of TAMs dynamics in shaping NAC response in BC. Notably, CD163+ TAMs may emerge as pivotal players in mechanisms of chemotherapy resistance and response, underscoring their potential as biomarkers and therapeutic targets in breast cancer treatment.
Collapse
Affiliation(s)
- Gizem Oner
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
- Department of General Surgery, Kocaeli University, Kocaeli, Turkey
| | | | - Hans Stoop
- Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | | | | | - Sevilay Altintas
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Christophe Van Berckelaer
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Zwi Berneman
- Department of Hematology, Antwerp University Hospital, Edegem, Belgium
| | - Wiebren Tjalma
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| | - Senada Koljenovic
- Department of Histopathology, Antwerp University Hospital, Edegem, Belgium
| | - Peter A van Dam
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
4
|
Lindberg I, Saleh A, Tutzauer J, Gunnarsdottir FB, Rydén L, Bergenfelz C, Larsson AM. Prognostic relevance of CD163 + immune cells in patients with metastatic breast cancer. Cancer Immunol Immunother 2025; 74:42. [PMID: 39751847 PMCID: PMC11699000 DOI: 10.1007/s00262-024-03892-2] [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: 08/15/2024] [Accepted: 11/11/2024] [Indexed: 01/04/2025]
Abstract
Metastatic breast cancer (MBC) is generally considered an incurable disease and even though new treatments are available, the median survival is approximately three years. The introduction of immune therapies for MBC highlights the importance of the immune system in cancer progression and treatment. CD163+ anti-inflammatory myeloid cells, including tumor associated macrophages (TAMs), are known to be of relevance in early breast cancer but their role in MBC is not yet established. Here we determine the levels of CD163+ immune cells in 139 patients with newly diagnosed MBC by using Immunohistochemistry (IHC) and gene expression analyses (GEX). We aim to determine changes and distribution of CD163+ immune cells during tumor progression from primary tumors (PT) to lymph node metastases (LNM) and distant metastases (DM). In addition, we evaluate associations between CD163+ immune cells, clinicopathological factors and disease outcome (progression-free and overall survival; PFS and OS, respectively). Despite similar distribution, high levels of CD163+ immune cells in the tumor nest of PT, but not in LNM or DM, associated with adverse prognostic features including higher grade and molecular subtype, as well as with shorter PFS and OS, however this observation was not significant after adjusted multivariate analyses. Finally, high levels of CD163+ immune cells in PT, as well as GEX in PT and synchronous LNM associated with shorter OS from the initial diagnosis. These results indicate that evaluating the levels of CD163+ immune cells may identify MBC patients with a worse prognosis. Unraveling the role of CD163+ immune cells in the complex immune responses in MBC is highly relevant for improving future immune therapies.
Collapse
Affiliation(s)
- Ida Lindberg
- Department of Translational Medicine, Division of Experimental Infection Medicine, Lund University, 221 84, Lund, Sweden
| | - Aya Saleh
- Department of Translational Medicine, Division of Experimental Infection Medicine, Lund University, 221 84, Lund, Sweden
| | - Julia Tutzauer
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 84, Lund, Sweden
- Department of Oncology and Pathology, Karolinska Institute and University Hospital, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Frida Björk Gunnarsdottir
- Department of Translational Medicine, Division of Experimental Infection Medicine, Lund University, 221 84, Lund, Sweden
| | - Lisa Rydén
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 84, Lund, Sweden
| | - Caroline Bergenfelz
- Department of Translational Medicine, Division of Experimental Infection Medicine, Lund University, 221 84, Lund, Sweden.
| | - Anna-Maria Larsson
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 84, Lund, Sweden
| |
Collapse
|
5
|
Hargrove-Wiley E, Obodo D, Bindeman W, Fingleton B. Elucidating Sex-Specific Immune Profiles in a Breast Cancer Model. Int J Mol Sci 2024; 25:13113. [PMID: 39684829 DOI: 10.3390/ijms252313113] [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: 10/30/2024] [Revised: 11/23/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Breast cancer is commonly thought of as a "women's disease". However, men are increasingly diagnosed with the disease, and their mortality rates are disparately higher than those of female patients. The abundance and composition of the immune microenvironment are determinants of breast cancer progression and survival. It is well documented that there are sex-specific differences in the immune response to several diseases, including various cancers. However, the effects of these differences in the context of breast cancer remain to be explored. This study demonstrates sex differences in the hormonal and immune landscape of the MMTV-PyMT transgenic murine model of female and male ER+ breast cancer using single-cell RNA sequencing (scRNA-Seq), whole-slide immunohistochemistry, and flow cytometry. Mammary tumors of transgenic male mice had increased estrogen receptor alpha expression and enriched nuclear binding signatures compared to female tumors. In the tumor immune compartment, male mice had lower intratumoral leukocyte infiltration. Yet, scRNA-Seq analysis reveals a more immunostimulatory microenvironment and increased antitumor immune populations in the primary and metastatic lungs as compared to transgenic females. Despite a more favorable innate immune profile, the metastatic burden was increased in male mice. Our data support a sex-dependent immune response in mammary carcinoma associated with the tumor, and likely host, hormonal environment. With emerging therapeutics targeting the tumor immune microenvironment, characterizing immune profiles is critical for optimizing their use in all breast cancer patients.
Collapse
Affiliation(s)
- Ebony Hargrove-Wiley
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Dora Obodo
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wendy Bindeman
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Barbara Fingleton
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
6
|
Fang C, Cheung MY, Chan RC, Poon IK, Lee C, To CC, Tsang JY, Li J, Tse GM. Prognostic Significance of CD163+ and/or CD206+ Tumor-Associated Macrophages Is Linked to Their Spatial Distribution and Tumor-Infiltrating Lymphocytes in Breast Cancer. Cancers (Basel) 2024; 16:2147. [PMID: 38893266 PMCID: PMC11172176 DOI: 10.3390/cancers16112147] [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: 04/26/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Tumor-associated macrophages (TAMs) is a key element in the breast tumor microenvironment. CD163 and CD206 have been utilized for TAM identification, but the clinical implications of TAMs identified by these markers have not been thoroughly explored. This study conducted a comparative analysis of CD163 and CD206 TAMs using digital image analysis, focusing on their spatial distribution and prognostic significance in relation to tumor-infiltrating lymphocytes (TILs). Distinct clinico-pathological and prognostic characteristics were noted between the two types of TAMs. CD163 TAMs were linked to high-grade tumors (p = 0.006), whereas CD206 TAMs were associated with a higher incidence of nodal metastasis (p = 0.033). CD206 TAMs were predominantly found in the stroma, with more cases being stromal CD206-high (sCD206-high) than tumoral CD206-high (tCD206-high) (p = 0.024). Regarding prognostication, patients stratified according to stromal and tumoral densities of CD163 showed different disease-free survival (DFS) time. Specifically, those that were sCD163-low but tCD163-high exhibited the poorest DFS (chi-square = 10.853, p = 0.013). Furthermore, a high sCD163-to-stromal-TILs ratio was identified as an independent predictor of unfavorable survival outcomes (DFS: HR = 3.477, p = 0.018). The spatial distribution and interactions with TILs enhanced the prognostic value of CD163 TAMs, while CD206 TAMs appeared to have limited prognostic utility in breast cancer cases.
Collapse
Affiliation(s)
- Canbin Fang
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Maisy Y. Cheung
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ronald C. Chan
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ivan K. Poon
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Conrad Lee
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Curtis C. To
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Julia Y. Tsang
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joshua Li
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Pathology, The University of Hong Kong, Hong Kong SAR, China
| | - Gary M. Tse
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Jääskeläinen MM, Tumelius R, Hämäläinen K, Rilla K, Oikari S, Rönkä A, Selander T, Mannermaa A, Tiainen S, Auvinen P. High Numbers of CD163+ Tumor-Associated Macrophages Predict Poor Prognosis in HER2+ Breast Cancer. Cancers (Basel) 2024; 16:634. [PMID: 38339385 PMCID: PMC10854814 DOI: 10.3390/cancers16030634] [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: 12/28/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are associated with a poor outcome in breast cancer (BC), but their prognostic value in different BC subtypes has remained somewhat unclear. Here, we investigated the prognostic value of M2-like TAMs (CD163+) and all TAMs (CD68+) in a patient cohort of 278 non-metastatic BC patients, half of whom were HER2+ (n = 139). The survival endpoints investigated were overall survival (OS), breast cancer-specific survival (BCSS) and disease-free survival (DFS). In the whole patient cohort (n = 278), a high CD163+ TAM count and a high CD68+ TAM count were associated with a worse outcome (p ≤ 0.023). In HER2+ BC, a high CD163+ TAM count was an independent factor for a poor prognosis across all the investigated survival endpoints (p < 0.001). The prognostic effect was evident in both the HER2+/hormone receptor-positive (p < 0.001) and HER2+/hormone receptor-negative (p ≤ 0.012) subgroups and regardless of the provision of adjuvant trastuzumab (p ≤ 0.002). In HER2-negative BC, the CD163+ TAM count was not significantly associated with survival. These results suggest that a high CD163+ TAM count predicts an inferior outcome, especially in HER2+ BC patients, and as adjuvant trastuzumab did not overcome the poor prognostic effect, combination treatments including therapies targeting the macrophage function could represent an effective therapeutic approach in HER2+ BC.
Collapse
Affiliation(s)
- Minna M. Jääskeläinen
- Cancer Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Ritva Tumelius
- Kuopio Center for Gene and Cell Therapy, 70210 Kuopio, Finland
| | - Kirsi Hämäläinen
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Imaging Center, Clinical Pathology, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Biocenter Kuopio and Cancer Center of Eastern Finland, University of Eastern Finland, 70211 Kuopio, Finland
| | - Kirsi Rilla
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Sanna Oikari
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Aino Rönkä
- Cancer Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Tuomas Selander
- Science Services Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
| | - Arto Mannermaa
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Wellbeing Services County of North Savo, 700029 Kuopio, Finland
| | - Satu Tiainen
- Cancer Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Päivi Auvinen
- Cancer Center, Kuopio University Hospital, Wellbeing Services County of North Savo, 70029 Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
8
|
Zhang M, Yue X, Zhao X, Lu Y, Liu H, Zhang Z, Ma H, Wang X, Xing H. Macrophage-specific deletion of Notch-1 induced M2 anti-inflammatory effect in atherosclerosis via activation of the PI3K-oxidative stress axis. Aging (Albany NY) 2023; 15:15196-15212. [PMID: 38149979 PMCID: PMC10781475 DOI: 10.18632/aging.205342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/02/2023] [Indexed: 12/28/2023]
Abstract
OBJECTIVE Notch-1 signaling is significantly associated with the occurrence and development of atherosclerosis (AS). However, the molecular mechanisms underlying the specific deletion of Notch-1 in AS-associated macrophages are not fully understood. This study aimed to investigate the effects of Notch-1 in AS. METHODS AND RESULTS Tissue samples were obtained from atherosclerotic segments of human carotid arteries. Immunofluorescence staining showed that Notch-1 was significantly colocalized with macrophages (CD68+), and Notch-1 staining was increased in human vulnerable plaques. Notch-1MAC-KO/ApoE-/- mice were generated in which Notch-1 was selectively inactivated in macrophages, and WT for littermate control mice (ApoE-/-/Notch-1WT). A control group was then established. All mice fed with a high-fat and Oil Red O, Movat, a-SMA, CD68, and Sirius red staining were used to evaluate the morphology. Specific deletion of Notch-1 in macrophages repressed the pathophysiology of AS. Immunofluorescent staining and Western blotting revealed that Notch-1MAC-KO repressed M1 and M2 responses in AS. Here, GSEA revealed that Notch-1 activation and PI3K signaling were statistically significantly correlated with each other, and Notch-1 was involved in the regulation of the PI3K signaling pathway. In the in vitro experiments, the secretion of Arg-1 and exosomes was classified by peritoneal macrophages of Notch-1MAC-KO/ApoE-/- and Notch-1WT/ApoE-/- mice. Immunohistochemistry staining and Western blotting were used to measure the expression levels of Notch1, PI3K, p-PI3K, AKT, p-AKT, Arg-1, IL-6, CD36, SREBP-1, CD206, iNOS, cleaved-caspase-3/-9, Bax, CD9, Alix and TSG101 in the peritoneal macrophages and exosomes, respectively. CONCLUSIONS The specific deletion of Notch-1 in macrophage represses the formation and development of AS via the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Mingming Zhang
- Clinical Medicine Research Center, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Xiangyong Yue
- Department of Oncology, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Xueping Zhao
- Department of Nursing, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Yonggang Lu
- Clinical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Hongtao Liu
- Clinical Medicine Research Center, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Zhe Zhang
- Clinical Medicine Research Center, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Huan Ma
- Clinical Medicine Research Center, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Xing Wang
- Clinical Medicine Research Center, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Hanying Xing
- Clinical Medicine Research Center, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| |
Collapse
|
9
|
Shao S, Miao H, Ma W. Unraveling the enigma of tumor-associated macrophages: challenges, innovations, and the path to therapeutic breakthroughs. Front Immunol 2023; 14:1295684. [PMID: 38035068 PMCID: PMC10682717 DOI: 10.3389/fimmu.2023.1295684] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are integral to the tumor microenvironment (TME), influencing cancer progression significantly. Attracted by cancer cell signals, TAMs exhibit unparalleled adaptability, aligning with the dynamic tumor milieu. Their roles span from promoting tumor growth and angiogenesis to modulating metastasis. While substantial research has explored the fundamentals of TAMs, comprehending their adaptive behavior, and leveraging it for novel treatments remains challenging. This review delves into TAM polarization, metabolic shifts, and the complex orchestration of cytokines and chemokines determining their functions. We highlight the complexities of TAM-targeted research focusing on their adaptability and potential variability in therapeutic outcomes. Moreover, we discuss the synergy of integrating TAM-focused strategies with established cancer treatments, such as chemotherapy, and immunotherapy. Emphasis is laid on pioneering methods like TAM reprogramming for cancer immunotherapy and the adoption of single-cell technologies for precision intervention. This synthesis seeks to shed light on TAMs' multifaceted roles in cancer, pinpointing prospective pathways for transformative research and enhancing therapeutic modalities in oncology.
Collapse
Affiliation(s)
- Shengwen Shao
- Clinical Research Center, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Huilai Miao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Hepatobiliary Surgery, Liaobu Hospital of Dongguan City, Dongguan, Guangdong, China
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center, and Sanford Stem Cell Institute, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
10
|
Lin J, Feng D, Liu J, Yang Y, Wei X, Lin W, Lin Q. Construction of stemness gene score by bulk and single-cell transcriptome to characterize the prognosis of breast cancer. Aging (Albany NY) 2023; 15:8185-8203. [PMID: 37602872 PMCID: PMC10496995 DOI: 10.18632/aging.204963] [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: 03/15/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
Breast cancer (BC) is a heterogeneous disease characterized by significant differences in prognosis and therapy response. Numerous prognostic tools have been developed for breast cancer. Usually these tools are based on bulk RNA-sequencing (RNA-Seq) and ignore tumor heterogeneity. Consequently, the goal of this study was to construct a single-cell level tool for predicting the prognosis of BC patients. In this study, we constructed a stemness-risk gene score (SGS) model based on single-sample gene set enrichment analysis (ssGSEA). Patients were divided into two groups based on the median SGS. Patients with a high SGS scores had a significantly worse prognosis than those with a low SGS, and these groups exhibited differences in several tumor characteristics, such as immune infiltration, gene mutations, and copy number variants. Our results indicate that the SGS is a reliable tool for predicting prognosis and response to immunotherapy in BC patients.
Collapse
Affiliation(s)
- Jun Lin
- Department of Anesthesiology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Anesthesiology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Deyi Feng
- Xiamen University, Xiamen 361100, China
| | - Jie Liu
- Department of Endoscopy, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Ye Yang
- The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xujin Wei
- The Graduate School of Fujian Medical University, Fuzhou 350001, China
| | - Wenqian Lin
- Department of Anesthesiology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Anesthesiology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Qun Lin
- Department of Anesthesiology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Anesthesiology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|