1
|
Ammarah U, Pereira‐Nunes A, Delfini M, Mazzone M. From monocyte-derived macrophages to resident macrophages-how metabolism leads their way in cancer. Mol Oncol 2024; 18:1739-1758. [PMID: 38411356 PMCID: PMC11223613 DOI: 10.1002/1878-0261.13618] [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: 10/23/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 02/28/2024] Open
Abstract
Macrophages are innate immune cells that play key roles during both homeostasis and disease. Depending on the microenvironmental cues sensed in different tissues, macrophages are known to acquire specific phenotypes and exhibit unique features that, ultimately, orchestrate tissue homeostasis, defense, and repair. Within the tumor microenvironment, macrophages are referred to as tumor-associated macrophages (TAMs) and constitute a heterogeneous population. Like their tissue resident counterpart, TAMs are plastic and can switch function and phenotype according to the niche-derived stimuli sensed. While changes in TAM phenotype are known to be accompanied by adaptive alterations in their cell metabolism, it is reported that metabolic reprogramming of macrophages can dictate their activation state and function. In line with these observations, recent research efforts have been focused on defining the metabolic traits of TAM subsets in different tumor malignancies and understanding their role in cancer progression and metastasis formation. This knowledge will pave the way to novel therapeutic strategies tailored to cancer subtype-specific metabolic landscapes. This review outlines the metabolic characteristics of distinct TAM subsets and their implications in tumorigenesis across multiple cancer types.
Collapse
Affiliation(s)
- Ummi Ammarah
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CentreUniversity of TorinoItaly
| | - Andreia Pereira‐Nunes
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
- Life and Health Sciences Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B's‐PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Marcello Delfini
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
| |
Collapse
|
2
|
Luo L, Deng S, Tang W, Hu X, Yin F, Ge H, Tang J, Liao Z, Feng J, Li X, Mo B. Monocytes subtypes from pleural effusion reveal biomarker candidates for the diagnosis of tuberculosis and malignancy. J Clin Lab Anal 2022; 36:e24579. [PMID: 35819097 PMCID: PMC9396188 DOI: 10.1002/jcla.24579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/08/2022] [Accepted: 06/18/2022] [Indexed: 11/22/2022] Open
Abstract
Background Pleural effusion is a common clinical condition caused by several respiratory diseases, including tuberculosis and malignancy. However, rapid and accurate diagnoses of tuberculous pleural effusion (TPE) and malignant pleural effusion (MPE) remain challenging. Although monocytes have been confirmed as an important immune cell in tuberculosis and malignancy, little is known about the role of monocytes subpopulations in the diagnosis of pleural effusion. Methods Pleural effusion samples and peripheral blood samples were collected from 40 TPE patients, 40 MPE patients, and 24 transudate pleural effusion patients, respectively. Chemokines (CCL2, CCL7, and CX3CL1) and cytokines (IL‐1β, IL‐17, IL‐27, and IFN‐γ) were measured by ELISA. The monocytes phenotypes were analyzed by flow cytometry. The chemokines receptors (CCR2 and CX3CR1) and cytokines above in different monocytes subsets were analyzed by real‐time PCR. Receiver operating characteristic curve analysis was performed for displaying differentiating power of intermediate and nonclassical subsets between tuberculous and malignant pleural effusions. Results CCL7 and CX3CL1 levels in TPE were significantly elevated in TPE compared with MPE and transudate pleural effusion. Cytokines, such as IL‐1β, IL‐17, IL‐27, and IFN‐γ, in TPE were much higher than in other pleural effusions. Moreover, CD14+CD16++ nonclassical subset frequency in TPE was remarkably higher than that in MPE, while CD14++CD16+ intermediate subset proportion in MPE was found elevated. Furthermore, CX3CL1‐CX3CR1 axis‐mediated infiltration of nonclassical monocytes in TPE was related to CX3CL1 and IFN‐γ expression in TPE. Higher expression of cytokines (IL‐1β, IL‐17, IL‐27, and IFN‐γ) were found in nonclassical monocytes compared with other subsets. Additionally, the proportions of intermediate and nonclassical monocytes in pleural effusion have the power in discriminating tuberculosis from malignant pleural effusion. Conclusions CD14 and CD16 markers on monocytes could be potentially used as novel diagnostic markers for diagnosing TPE and MPE.
Collapse
Affiliation(s)
- Lisha Luo
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Shuanglinzi Deng
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Tang
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Xinyue Hu
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Feifei Yin
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Huan Ge
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Jiale Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhonghua Liao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Juntao Feng
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaozhao Li
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Biwen Mo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.,Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, Guilin, China
| |
Collapse
|
3
|
Ozkan E, Bakar-Ates F. The Trinity of Matrix Metalloproteinases, Inflammation, and Cancer: A Literature Review of Recent Updates. Antiinflamm Antiallergy Agents Med Chem 2021; 19:206-221. [PMID: 32178620 PMCID: PMC7499348 DOI: 10.2174/1871523018666191023141807] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/02/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
The critical link between cancer and inflammation has been known for many years. This complex network was further complexed by revealing the association of the matrix metalloproteinase family members with inflammatory cytokines, which were previously known to be responsible for the development of metastasis. This article summarizes the current studies which evaluate the relationship between cancer and inflammatory microenvironment as well as the roles of MMPs on invasion and metastasis together.
Collapse
Affiliation(s)
- Erva Ozkan
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Filiz Bakar-Ates
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
4
|
Macrophage Polarization States in the Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22136995. [PMID: 34209703 PMCID: PMC8268869 DOI: 10.3390/ijms22136995] [Citation(s) in RCA: 928] [Impact Index Per Article: 232.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/09/2021] [Accepted: 06/25/2021] [Indexed: 12/13/2022] Open
Abstract
The M1/M2 macrophage paradigm plays a key role in tumor progression. M1 macrophages are historically regarded as anti-tumor, while M2-polarized macrophages, commonly deemed tumor-associated macrophages (TAMs), are contributors to many pro-tumorigenic outcomes in cancer through angiogenic and lymphangiogenic regulation, immune suppression, hypoxia induction, tumor cell proliferation, and metastasis. The tumor microenvironment (TME) can influence macrophage recruitment and polarization, giving way to these pro-tumorigenic outcomes. Investigating TME-induced macrophage polarization is critical for further understanding of TAM-related pro-tumor outcomes and potential development of new therapeutic approaches. This review explores the current understanding of TME-induced macrophage polarization and the role of M2-polarized macrophages in promoting tumor progression.
Collapse
|
5
|
Sedighzadeh SS, Khoshbin AP, Razi S, Keshavarz-Fathi M, Rezaei N. A narrative review of tumor-associated macrophages in lung cancer: regulation of macrophage polarization and therapeutic implications. Transl Lung Cancer Res 2021; 10:1889-1916. [PMID: 34012800 PMCID: PMC8107755 DOI: 10.21037/tlcr-20-1241] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer is the deadliest malignancy worldwide. An inflammatory microenvironment is a key factor contributing to lung tumor progression. Tumor-Associated Macrophages (TAMs) are prominent components of the cancer immune microenvironment with diverse supportive and inhibitory effects on growth, progression, and metastasis of lung tumors. Two main macrophage phenotypes with different functions have been identified. They include inflammatory or classically activated (M1) and anti-inflammatory or alternatively activated (M2) macrophages. The contrasting functions of TAMs in relation to lung neoplasm progression stem from the presence of TAMs with varying tumor-promoting or anti-tumor activities. This wide spectrum of functions is governed by a network of cytokines and chemokines, cell-cell interactions, and signaling pathways. TAMs are promising therapeutic targets for non-small cell lung cancer (NSCLC) treatment. There are several strategies for TAM targeting and utilizing them for therapeutic purposes including limiting monocyte recruitment and localization through various pathways such as CCL2-CCR2, CSF1-CSF1R, and CXCL12-CXCR4, targeting the activation of TAMs, genetic and epigenetic reprogramming of TAMs to antitumor phenotype, and utilizing TAMs as the carrier for anti-cancer drugs. In this review, we will outline the role of macrophages in the lung cancer initiation and progression, pathways regulating their function in lung cancer microenvironment as well as the role of these immune cells in the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Sahar Sadat Sedighzadeh
- Department of Biological Sciences, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amin Pastaki Khoshbin
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Sheffield, UK
| |
Collapse
|
6
|
Martín-Sierra C, Martins R, Coucelo M, Abrantes AM, Oliveira RC, Tralhão JG, Botelho MF, Furtado E, Domingues MR, Paiva A, Laranjeira P. Elevated soluble TNFα levels and upregulated TNFα mRNA expression in purified peripheral blood monocyte subsets associated with high-grade hepatocellular carcinoma. J Inflamm (Lond) 2020; 17:14. [PMID: 32256215 PMCID: PMC7106708 DOI: 10.1186/s12950-020-00243-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/13/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Chronic inflammation is involved in the initiation and progression of various cancers, including liver cancer. The current study focuses on the characterization of the peripheral immune response in hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) patients, before and after surgical procedure, in order to assess the effect of tumor resection in the immune system homeostasis and to determine possible prognostic factors associated with high-grade tumors. We developed a whole-blood assay to monitor immune alterations and functional competence of peripheral monocytes in a group of 10 healthy individuals (HG), in 20 HCC patients and 8 CCA patients, by multi-color flow cytometry, qRT-PCR, and ELISA techniques. RESULTS The qRT-PCR analysis showed an upregulation of TNFα expression by classical and intermediate monocytes purified from HCC patients presenting tumors in grade G3-G4 as compared to G1-G2 HCC patients. Moreover, ELISA assay confirmed elevated serum levels of TNFα in G3-G4 compared to G1-G2 HCC patients. A significant decrease of circulating non-classical monocytes was detected in both CCA and HCC patients before and after surgical procedure. In addition, a functional defect in circulating classical and intermediate monocytes was observed in both groups of cancer patients when compared to the HG, with partial recovery after the surgical intervention. CONCLUSIONS This integrated analysis permitted the identification of altered functional competence of monocyte subsets in CCA and HCC patients. In addition, our results point to a potential role of TNFα as a prognostic peripheral biomarker in HCC patients, indicating the presence of high-grade tumors that should be further validated.
Collapse
Affiliation(s)
- C. Martín-Sierra
- Flow Cytometry Unit, Clinical Pathology Service, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, Ed. S. Jerónimo, 3° piso, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - R. Martins
- Unidade Transplantação Hepática Pediátrica e de Adultos, Centro Hospitalar e Universitário de Coimbra (UTHPA, CHUC), Coimbra, Portugal
- Serviço de Cirurgia Geral, Unidade HBP, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- Instituto de Biofísica, IBILI, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - M. Coucelo
- Unidade de Hematologia Molecular, Serviço de Hematologia Clínica, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - A. M. Abrantes
- Instituto de Biofísica, IBILI, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - R. C. Oliveira
- Instituto de Biofísica, IBILI, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Serviço de Anatomia Patológica, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - J. G. Tralhão
- Unidade Transplantação Hepática Pediátrica e de Adultos, Centro Hospitalar e Universitário de Coimbra (UTHPA, CHUC), Coimbra, Portugal
- Serviço de Cirurgia Geral, Unidade HBP, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
- Instituto de Biofísica, IBILI, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - M. F. Botelho
- Instituto de Biofísica, IBILI, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - E. Furtado
- Unidade Transplantação Hepática Pediátrica e de Adultos, Centro Hospitalar e Universitário de Coimbra (UTHPA, CHUC), Coimbra, Portugal
| | - M. R. Domingues
- Mass Spectrometry Centre, Department of Chemistry & QOPNA, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - A. Paiva
- Flow Cytometry Unit, Clinical Pathology Service, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, Ed. S. Jerónimo, 3° piso, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Coimbra, Portugal
| | - P. Laranjeira
- Flow Cytometry Unit, Clinical Pathology Service, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, Ed. S. Jerónimo, 3° piso, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
7
|
Bleomycin inhibits proliferation and induces apoptosis in TPC-1 cells through reversing M2-macrophages polarization. Oncol Lett 2018; 16:3858-3866. [PMID: 30127999 PMCID: PMC6096247 DOI: 10.3892/ol.2018.9103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 07/03/2018] [Indexed: 12/17/2022] Open
Abstract
Papillary thyroid carcinoma (PTC) is one of the most common types of thyroid malignancy. Previous studies have demonstrated that the density of tumor-associated macrophages (TAMs) within the tumor microenvironment affects the progression of PTC due to the imbalance in M1/M2 macrophage subtypes. M2 macrophages induce anti-inflammatory effects and promote tumor progression, whereas M1 macrophages destroy tumor cells. Therefore, reversing TAM polarization to M1 may be a novel strategy for the treatment of cancer. Although bleomycin (BLM) is a commonly used anti-cancer drug, which regulates the secretion of relevant cytokines, high dose and long-term treatment with BLM may lead to pulmonary fibrosis. In the present study, flow cytometry data revealed that low dose treatment with BLM (5 or 10 mU/ml) facilitated the expression of the M1 phenotype markers cluster of differentiation (CD)80 and C-C chemokine receptor 7, and concurrently suppressed the M2 marker CD206 on M2-macrophages. Reverse transcription-quantitative polymerase chain reaction data revealed that the expression levels of tumor necrosis factor-α and interleukin-1β markedly increased, whereas the expression of IL-10 decreased in M2 macrophages treated with BLM. A fluorescein isothiocyanate-dextran uptake experiment revealed that BLM increased the phagocytic capacity of M2, however not M1 or M0 macrophages. In addition, to verify the effect of BLM-treated M2 macrophages on thyroid carcinoma cells, a co-culture system of macrophages and the human PTC cell line TPC-1, was established. BLM-treated M2 macrophages increased the number of cells in early and late apoptosis and inhibited the migration, proliferation and invasion of TPC-1 cells. These results suggest that a low dose and indirect effect of BLM may induce suppressive effects on PTC by selectively reversing M2 macrophage polarization to M1, which may provide a novel strategy for cancer treatment.
Collapse
|
8
|
Narasimhan PB, Akabas L, Tariq S, Huda N, Bennuru S, Sabzevari H, Hofmeister R, Nutman TB, Tolouei Semnani R. Similarities and differences between helminth parasites and cancer cell lines in shaping human monocytes: Insights into parallel mechanisms of immune evasion. PLoS Negl Trop Dis 2018; 12:e0006404. [PMID: 29668679 PMCID: PMC5927465 DOI: 10.1371/journal.pntd.0006404] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/30/2018] [Accepted: 03/22/2018] [Indexed: 12/24/2022] Open
Abstract
A number of features at the host-parasite interface are reminiscent of those that are also observed at the host-tumor interface. Both cancer cells and parasites establish a tissue microenvironment that allows for immune evasion and may reflect functional alterations of various innate cells. Here, we investigated how the phenotype and function of human monocytes is altered by exposure to cancer cell lines and if these functional and phenotypic alterations parallel those induced by exposure to helminth parasites. Thus, human monocytes were exposed to three different cancer cell lines (breast, ovarian, or glioblastoma) or to live microfilariae (mf) of Brugia malayi-a causative agent of lymphatic filariasis. After 2 days of co-culture, monocytes exposed to cancer cell lines showed markedly upregulated expression of M1-associated (TNF-α, IL-1β), M2-associated (CCL13, CD206), Mreg-associated (IL-10, TGF-β), and angiogenesis associated (MMP9, VEGF) genes. Similar to cancer cell lines, but less dramatically, mf altered the mRNA expression of IL-1β, CCL13, TGM2 and MMP9. When surface expression of the inhibitory ligands PDL1 and PDL2 was assessed, monocytes exposed to both cancer cell lines and to live mf significantly upregulated PDL1 and PDL2 expression. In contrast to exposure to mf, exposure to cancer cell lines increased the phagocytic ability of monocytes and reduced their ability to induce T cell proliferation and to expand Granzyme A+ CD8+ T cells. Our data suggest that despite the fact that helminth parasites and cancer cell lines are extraordinarily disparate, they share the ability to alter the phenotype of human monocytes.
Collapse
Affiliation(s)
- Prakash Babu Narasimhan
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Leor Akabas
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Sameha Tariq
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Naureen Huda
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Sasisekhar Bennuru
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Helen Sabzevari
- EMD Serono Research and Development Institute, Billerica, MA, United States of America
| | - Robert Hofmeister
- EMD Serono Research and Development Institute, Billerica, MA, United States of America
| | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Roshanak Tolouei Semnani
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
9
|
Ke X, Zhang S, Wu M, Lou J, Zhang J, Xu T, Huang L, Huang P, Wang F, Pan S. Tumor-associated macrophages promote invasion via Toll-like receptors signaling in patients with ovarian cancer. Int Immunopharmacol 2016; 40:184-195. [PMID: 27608303 DOI: 10.1016/j.intimp.2016.08.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/21/2016] [Accepted: 08/24/2016] [Indexed: 12/17/2022]
Abstract
Tumor-associated macrophages (TAMs) derived from peripheral blood monocytes recruit into tumor microenvironment and display functions associated with tumor progression. The mechanisms by which TAMs display roles that associated with the invasion ability of ovarian cancer have not been well investigated. In our research, we found abundant TAMs infiltrate in ovarian cancer compared with benign ovarian tumor tissues. Levels of matrix metalloproteinase (MMP)-2, MMP-9 and MMP-10, and Toll-like receptors (TLRs) signaling proteins were evaluated in ovarian cancer. The high level of TAMs was associated with metastasis and advance of patients with ovarian cancer. TAMs and ovarian cancer cell line SKOV3 were cocultured in vitro, MMPs level and the invasion ability of SKOV3 cells were significantly up-regulated. The coculture process was correlated with the activation of TLRs signaling and downstream nuclear factor (NF)-κB p65 and microtubule-associated proteins (MAPs) kinases pathway in SKOV3. In addition, pre-incubation with TLRs signaling inhibitors remarkably suppressed invasion ability of SKOV3. Levels of TLRs signaling pathways proteins were also down-regulated in this blocking process. These findings demonstrated that TAMs promoted up-regulation of MMP-2, MMP-9 and MMP-10 expressions and enhanced ovarian cancer cells invasion via TLRs signaling pathway. We conclude that TAMs could enhance ovarian cancer cells invasion and ultimately promote ovarian cancer progression.
Collapse
Affiliation(s)
- Xing Ke
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China
| | - Shuping Zhang
- Department of Laboratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, 210029 Nanjing, China
| | - Meng Wu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China
| | - Jianfang Lou
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China
| | - Jiexin Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China
| | - Ting Xu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China
| | - Lei Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China
| | - Peijun Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China.
| | - Shiyang Pan
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China.
| |
Collapse
|
10
|
Interplay between Cellular and Molecular Inflammatory Mediators in Lung Cancer. Mediators Inflamm 2016; 2016:3494608. [PMID: 26941482 PMCID: PMC4749813 DOI: 10.1155/2016/3494608] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 01/10/2016] [Indexed: 01/09/2023] Open
Abstract
Inflammation is a component of the tumor microenvironment and represents the 7th hallmark of cancer. Chronic inflammation plays a critical role in tumorigenesis. Tumor infiltrating inflammatory cells mediate processes associated with progression, immune suppression, promotion of neoangiogenesis and lymphangiogenesis, remodeling of extracellular matrix, invasion and metastasis, and, lastly, the inhibition of vaccine-induced antitumor T cell response. Accumulating evidence indicates a critical role of myeloid cells in the pathophysiology of human cancers. In contrast to the well-characterized tumor-associated macrophages (TAMs), the significance of granulocytes in cancer has only recently begun to emerge with the characterization of tumor-associated neutrophils (TANs). Recent studies show the importance of CD47 in the interaction with macrophages inhibiting phagocytosis and promoting the migration of neutrophils, increasing inflammation which can lead to recurrence and progression in lung cancer. Currently, therapies are targeted towards blocking CD47 and enhancing macrophage-mediated phagocytosis. However, antibody-based therapies may have adverse effects that limit its use.
Collapse
|
11
|
Verronèse E, Delgado A, Valladeau-Guilemond J, Garin G, Guillemaut S, Tredan O, Ray-Coquard I, Bachelot T, N'Kodia A, Bardin-Dit-Courageot C, Rigal C, Pérol D, Caux C, Ménétrier-Caux C. Immune cell dysfunctions in breast cancer patients detected through whole blood multi-parametric flow cytometry assay. Oncoimmunology 2015; 5:e1100791. [PMID: 27141361 PMCID: PMC4839376 DOI: 10.1080/2162402x.2015.1100791] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/18/2015] [Accepted: 09/22/2015] [Indexed: 01/08/2023] Open
Abstract
Monitoring functional competence of immune cell populations in clinical routine represents a major challenge. We developed a whole-blood assay to monitor functional competence of peripheral innate immune cells including NK cells, dendritic and monocyte cell subsets through their ability to produce specific cytokines after short-term stimulation, detected through intra-cytoplasmic staining and multi-parametric flow-cytometry. A PMA/ionomycin T cell activation assay complemented this analysis. Comparing cohorts of healthy women and breast cancer (BC) patients at different stages, we identified significant functional alteration of circulating immune cells during BC progression prior to initiation of treatment. Of upmost importance, as early as the localized primary tumor (PT) stage, we observed functional alterations in several innate immune populations and T cells i.e. (i) reduced TNFα production by BDCA-1+ DC and non-classical monocytes in response to Type-I IFN, (ii) a strong drop in IFNγ production by NK cells in response to either Type-I IFN or TLR7/8 ligand, and (iii) a coordinated impairment of cytokine (IL-2, IFNγ, IL-21) production by T cell subpopulations. Overall, these alterations are further accentuated according to the stage of the disease in first-line metastatic patients. Finally, whereas we did not detect functional modification of DC subsets in response to TLR7/8 ligand, we highlighted increased IL-12p40 production by monocytes specifically at first relapse (FR). Our results reinforce the importance of monitoring both innate and adaptive immunity to better evaluate dysfunctions in cancer patients and suggest that our whole-blood assay will be useful to monitor response to treatment, particularly for immunotherapeutic strategies.
Collapse
Affiliation(s)
- E Verronèse
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center , Lyon, France
| | - A Delgado
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center , Lyon, France
| | - J Valladeau-Guilemond
- Team 11, INSERM U1052/CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France
| | - G Garin
- DRCI department, Léon Bérard Cancer Center , Lyon, France
| | - S Guillemaut
- DRCI department, Léon Bérard Cancer Center , Lyon, France
| | - O Tredan
- Department of Medical Oncology, Léon Bérard Cancer Center , Lyon, France
| | - I Ray-Coquard
- Department of Medical Oncology, Léon Bérard Cancer Center , Lyon, France
| | - T Bachelot
- Team 11, INSERM U1052/CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; Department of Medical Oncology, Léon Bérard Cancer Center, Lyon, France
| | - A N'Kodia
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center , Lyon, France
| | - C Bardin-Dit-Courageot
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center , Lyon, France
| | - C Rigal
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center , Lyon, France
| | - D Pérol
- DRCI department, Léon Bérard Cancer Center , Lyon, France
| | - C Caux
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center, Lyon, France; Team 11, INSERM U1052/CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France
| | - C Ménétrier-Caux
- Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Léon Bérard Cancer Center, Lyon, France; Team 11, INSERM U1052/CNRS UMR5286, Cancer Research Center of Lyon, Lyon, France; Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France
| |
Collapse
|
12
|
Almatroodi SA, McDonald CF, Darby IA, Pouniotis DS. Characterization of M1/M2 Tumour-Associated Macrophages (TAMs) and Th1/Th2 Cytokine Profiles in Patients with NSCLC. CANCER MICROENVIRONMENT 2015; 9:1-11. [PMID: 26319408 DOI: 10.1007/s12307-015-0174-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
Abstract
Lung cancer is one of the most commonly reported cancers, and is known to be associated with a poor prognosis. The function of tumour-associated macrophages (TAMs) in lung cancer patients is multifaceted and the literature shows conflicting roles. (I) To analyze the Th1 and Th2 cytokine levels that contribute to the differentiation of M1 and M2 macrophage populations in the serum of patients with NSCLC versus non-cancer controls; and (II) To characterize the M1 and M2 macrophage populations within TAMs in different subtypes of NSCLC compared to non-tumour tissue. The Th1 and Th2 cytokine levels were analyzed in serum using the Bio-Plex assay. In addition, TAMs subsets from non-tumour and tumour tissues were analyzed using immunohistochemistry (IHC). The level of IL-1β, IL-4, IL-6 and IL-8 was found to be increased in the serum of patients with large cell carcinoma but not in other NSCLC subtypes compared to non-cancer controls. In addition, the expression of CD68 and M2 marker CD163 was found to be increased (P ≤ 0.0001) in all NSCLC subtypes compared to non-tumour tissues. In contrast, the expression of iNOS (M1 marker) was decreased in the tumour tissue of patients with adenocarcinoma (P ≤ 0.01) and squamous carcinoma (P ≤ 0.05) but not in large cell carcinoma compared to non-tumour tissue. The results of this study indicate that NSCLC might have the ability to alter phenotype within the lung tumour areas in the local environment (TAMs) but not in the bloodstream in the systemic environment (serum) except for large cell carcinoma.
Collapse
Affiliation(s)
- S A Almatroodi
- Cancer & Tissue Repair Laboratory, School of Medical Sciences, RMIT University, P.O. Box 71, Bundoora, Victoria, Australia, 3083. .,Applied Medical Sciences College, Qassim University, Buraidah, Saudi Arabia.
| | - C F McDonald
- Institute for Breathing & Sleep, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - I A Darby
- Cancer & Tissue Repair Laboratory, School of Medical Sciences, RMIT University, P.O. Box 71, Bundoora, Victoria, Australia, 3083
| | - D S Pouniotis
- Cancer & Tissue Repair Laboratory, School of Medical Sciences, RMIT University, P.O. Box 71, Bundoora, Victoria, Australia, 3083
| |
Collapse
|
13
|
Alveolar Macrophage Polarisation in Lung Cancer. LUNG CANCER INTERNATIONAL 2014; 2014:721087. [PMID: 26316944 PMCID: PMC4437403 DOI: 10.1155/2014/721087] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/11/2014] [Indexed: 01/03/2023]
Abstract
The role of alveolar macrophages in lung cancer is multifaceted and conflicting. Alveolar macrophage secretion of proinflammatory cytokines has been found to enhance antitumour functions, cytostasis (inhibition of tumour growth), and cytotoxicity (macrophage-mediated killing). In contrast, protumour functions of alveolar macrophages in lung cancer have also been indicated. Inhibition of antitumour function via secretion of the anti-inflammatory cytokine IL-10 as well as reduced secretion of proinflammatory cytokines and reduction of mannose receptor expression on alveolar macrophages may contribute to lung cancer progression and metastasis. Alveolar macrophages have also been found to contribute to angiogenesis and tumour growth via the secretion of IL-8 and VEGF. This paper reviews the evidence for a dual role of alveolar macrophages in lung cancer progression.
Collapse
|
14
|
Huang JJ, Li YJ, Xia Y, Wang Y, Wei WX, Zhu YJ, Lin TY, Huang HQ, Jiang WQ, Li ZM. Prognostic significance of peripheral monocyte count in patients with extranodal natural killer/T-cell lymphoma. BMC Cancer 2013; 13:222. [PMID: 23638998 PMCID: PMC3653743 DOI: 10.1186/1471-2407-13-222] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 04/25/2013] [Indexed: 01/02/2023] Open
Abstract
Background Extranodal natural killer/T-cell lymphoma (ENKL) has heterogeneous clinical manifestations and prognosis. This study aims to evaluate the prognostic impact of absolute monocyte count (AMC) in ENKL, and provide some immunologically relevant information for better risk stratification in patients with ENKL. Methods Retrospective data from 163 patients newly diagnosed with ENKL were analyzed. The absolute monocyte count (AMC) at diagnosis was analyzed as continuous and dichotomized variables. Independent prognostic factors of survival were determined by Cox regression analysis. Results The AMC at diagnosis were related to overall survival (OS) and progression-free survival (PFS) in patients with ENKL. Multivariate analysis identified AMC as independent prognostic factors of survival, independent of International Prognostic Index (IPI) and Korean prognostic index (KPI). The prognostic index incorporating AMC and absolute lymphocyte count (ALC), another surrogate factor of immune status, could be used to stratify all 163 patients with ENKL into different prognostic groups. For patients who received chemotherapy followed by radiotherapy (102 cases), the three AMC/ALC index categories identified patients with significantly different survivals. When superimposed on IPI or KPI categories, the AMC/ALC index was better able to identify high-risk patients in the low-risk IPI or KPI category. Conclusion The baseline peripheral monocyte count is shown to be an effective prognostic indicator of survival in ENKL patients. The prognostic index related to tumor microenvironment might be helpful to identify high-risk patients with ENKL.
Collapse
Affiliation(s)
- Jia-Jia Huang
- State Key Laboratory of Oncology in South China, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Squamous carcinoma cells influence monocyte phenotype and suppress lipopolysaccharide-induced TNF-alpha in monocytes. Inflammation 2010; 33:207-23. [PMID: 20084448 DOI: 10.1007/s10753-009-9175-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bacteria and chronic inflammation are present in squamous cell carcinoma of the head and neck (HNSCC), but their roles in the pathogenesis of HNSCC are unclear. Our studies described here revealed that human monocytes co-cultured short term with HNSCC cells were more likely to express CD16, and CD16(+) small mononuclear cells were common in HNSCC specimens. In addition, we identified monocytes as the primary source of LPS-induced IL-6 and TNF-alpha in the monocyte-HNSCC co-cultures. Remarkably, relative to LPS-stimulated monocytes cultured alone, HNSCC cells profoundly suppressed LPS-induced TNF-alpha in monocytes, without compromising IL-6 production. High levels of cytoprotective factors like IL-6 and low levels of TNF-alpha are important for the tumor microenvironment that enables tumor cell survival, affects monocyte differentiation and may contribute to tumor colonization by bacteria. This study provides novel observations that HNSCC cells affect monocyte phenotype and function, which are relevant to the regulation of the HNSCC microenvironment.
Collapse
|
16
|
Richardt-Pargmann D, Wechsler M, Krieg AM, Vollmer J, Jurk M. Positive T cell co-stimulation by TLR7/8 ligands is dependent on the cellular environment. Immunobiology 2010; 216:12-23. [PMID: 20542588 DOI: 10.1016/j.imbio.2010.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 03/12/2010] [Accepted: 03/25/2010] [Indexed: 11/30/2022]
Abstract
Toll-like receptors (TLRs) are mediators of innate immune responses detecting conserved pathogen-associated molecules. Whereas most TLRs are expressed on the cell surface, TLR3, 7, 8 and 9 are predominantly localized in endosomal compartments. Recent studies reported that TLRs are also expressed by T lymphocytes, resulting in direct co-stimulation of isolated CD4(+) T cells for example by Pam3CSK4 (TLR2 ligand) or flagellin (TLR5 ligand). We here describe enhanced IFN-γ production and T cell proliferation by anti-CD3 T cell receptor (TCR) or antigenic stimulation of purified human CD4(+) T cells upon co-culture with TLR7/8 specific single-stranded oligoribonucleotides or small molecule ligands. Surprisingly, TLR7/8 stimulation of CD4(+) T cells within a whole peripheral mononuclear cell (PBMC) environment did not result in enhanced T cell proliferation, but in a lack of proliferation that was cell-cell contact dependent. Immune cell depletion assays pointed towards a monocyte-mediated effect. Different TLR ligands influenced T cell proliferation differently. The effect of inhibition of T cell proliferation was most prominently seen for TLR7 ligands whereas the effects were minimal for TLR8 and TLR9 ligands indicating that the suppressive phenotype is unique only for certain TLRs. Our results strongly suggest that co-stimulation of T cell proliferation by TLR7/8 agonists is dependent on the specific cellular context.
Collapse
Affiliation(s)
- Denise Richardt-Pargmann
- Pfizer Oligonucleotide Therapeutics Unit (OTU), Coley Pharmaceutical GmbH, Merowingerplatz, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|