1
|
Canè S, Geiger R, Bronte V. The roles of arginases and arginine in immunity. Nat Rev Immunol 2024:10.1038/s41577-024-01098-2. [PMID: 39420221 DOI: 10.1038/s41577-024-01098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Arginase activity and arginine metabolism in immune cells have important consequences for health and disease. Their dysregulation is commonly observed in cancer, autoimmune disorders and infectious diseases. Following the initial description of a role for arginase in the dysfunction of T cells mounting an antitumour response, numerous studies have broadened our understanding of the regulation and expression of arginases and their integration with other metabolic pathways. Here, we highlight the differences in arginase compartmentalization and storage between humans and rodents that should be taken into consideration when assessing the effects of arginase activity. We detail the roles of arginases, arginine and its metabolites in immune cells and their effects in the context of cancer, autoimmunity and infectious disease. Finally, we explore potential therapeutic strategies targeting arginases and arginine.
Collapse
Affiliation(s)
- Stefania Canè
- The Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Roger Geiger
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Università della Svizzera italiana, Bellinzona, Switzerland
| | | |
Collapse
|
2
|
Tang X, Liu W, Liang J, Zhu X, Ge X, Fang D, Ling L, Yuan F, Zeng K, Chen Q, Zhang G, Gong L, Zhang S. Triamcinolone Acetonide Protects Against Light-Induced Retinal Degeneration by Activating Anti-Inflammatory STAT6/Arg1 Signaling in Microglia. Inflammation 2024:10.1007/s10753-024-02152-w. [PMID: 39340587 DOI: 10.1007/s10753-024-02152-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Affiliation(s)
- Xiangcheng Tang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, JinanUniversity, 18 Zetian Road, Shenzhen, 518040, Guangdong, China
| | - Wei Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China
| | - Jia Liang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, JinanUniversity, 18 Zetian Road, Shenzhen, 518040, Guangdong, China
| | - Xingfei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China
| | - Xiangyu Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China
| | - Dong Fang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, JinanUniversity, 18 Zetian Road, Shenzhen, 518040, Guangdong, China
| | - Lirong Ling
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China
| | - Fanglan Yuan
- Shenzhen Eye Hospital, Shenzhen Eye Institute, JinanUniversity, 18 Zetian Road, Shenzhen, 518040, Guangdong, China
| | - Kun Zeng
- Shenzhen Eye Hospital, Shenzhen Eye Institute, JinanUniversity, 18 Zetian Road, Shenzhen, 518040, Guangdong, China
| | - Qingshan Chen
- Shenzhen Eye Hospital, Shenzhen Eye Institute, JinanUniversity, 18 Zetian Road, Shenzhen, 518040, Guangdong, China
| | - Guoming Zhang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, JinanUniversity, 18 Zetian Road, Shenzhen, 518040, Guangdong, China
| | - Lili Gong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, Guangdong, China.
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510230, Guangdong, China.
| | - Shaochong Zhang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, JinanUniversity, 18 Zetian Road, Shenzhen, 518040, Guangdong, China.
| |
Collapse
|
3
|
Mizuno N, Shiga S, Tanaka Y, Kimura T, Yanagawa Y. CDK8/19 inhibitor enhances arginase-1 expression in macrophages via STAT6 and p38 MAPK activation. Eur J Pharmacol 2024; 979:176852. [PMID: 39067565 DOI: 10.1016/j.ejphar.2024.176852] [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: 03/24/2024] [Revised: 06/25/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Macrophages polarize into alternatively activated M2 macrophages through interleukin (IL)-4, and they express high levels of arginase-1, which promotes anti-inflammatory responses. Several studies have confirmed the anti-inflammatory effects of cyclin-dependent kinase (CDK) 8/19 inhibition, and hence, numerous CDK8/19 inhibitors, such as BRD6989, have been developed. However, the effects of CDK8/19 inhibitors on arginase-1 expression in macrophages have not yet been elucidated. This study investigated the effects of CDK8/19 inhibitor on arginase-1 expression in IL-4-activated macrophages. The results showed that BRD6989 increased arginase-1 expression transcriptionally in murine peritoneal macrophages and the murine macrophage cell line RAW264.7 in an IL-4-dependent manner. In addition, the results indicated that BRD6989 enhances signal transducer and activator of transcription (STAT) 6 phosphorylation. Meanwhile, BRD6989 exhibited the capability to activate p38 mitogen-activated protein kinase (MAPK) even in the absence of IL-4 stimulation. Moreover, we observed that a p38 MAPK inhibitor suppressed the BRD6989-induced increase in arginase-1 expression. Besides, BRD6989 increased the surface expression of CD206, an M2 macrophage marker. Thus, this study demonstrated for the first time that CDK8/19 inhibition increases arginase-1 expression, suggesting that this mechanism involves the activation of STAT6 and p38 MAPK. This finding implies that CDK8/19 inhibition may facilitate the production of anti-inflammatory M2 macrophages.
Collapse
Affiliation(s)
- Natsumi Mizuno
- Department of Pharmacology, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, Tobetsu, Ishikari, 061-0293, Japan.
| | - Saki Shiga
- Department of Pharmacology, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, Tobetsu, Ishikari, 061-0293, Japan
| | - Yoshiyuki Tanaka
- Department of Pharmacology, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, Tobetsu, Ishikari, 061-0293, Japan
| | - Tatsuki Kimura
- Department of Pharmacology, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, Tobetsu, Ishikari, 061-0293, Japan
| | - Yoshiki Yanagawa
- Department of Pharmacology, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Kanazawa 1757, Tobetsu, Ishikari, 061-0293, Japan
| |
Collapse
|
4
|
Gu Q, Qi A, Wang N, Zhou Z, Zhou X. Macrophage dynamics in prostate cancer: Molecular to therapeutic insights. Biomed Pharmacother 2024; 177:117002. [PMID: 38960836 DOI: 10.1016/j.biopha.2024.117002] [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: 04/09/2024] [Revised: 06/08/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024] Open
Abstract
This review provides an in-depth examination of the role that tumor-associated macrophages (TAMs) play in the progression of prostate cancer (PCa), with a particular focus on the factors influencing the polarization of M1 and M2 macrophages and the implications of targeting these cells for cancer progression. The development and prognosis of PCa are significantly influenced by the behavior of macrophages within the tumor microenvironment. M1 macrophages typically exhibit anti-tumor properties by secreting pro-inflammatory cytokines such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), thereby enhancing the immune response. Conversely, M2 macrophages contribute to tumor cell migration and invasion through the production of factors like arginase-1 (Arg1) and interleukin-10 (IL-10). This review not only explores the diverse factors that affect macrophage polarization but also delves into the potential therapeutic strategies targeting macrophage polarization, including the critical roles of non-coding RNA and exosomes in regulating this process. The polarization state of macrophages is highlighted as a key determinant in PCa progression, offering a novel perspective for clinical treatment. Future research should concentrate on gaining a deeper understanding of the molecular mechanisms underlying macrophage polarization and on developing effective targeted therapeutic strategies. The exploration of the potential of combination therapies to improve treatment efficacy is also emphasized. By emphasizing the importance of macrophages as a therapeutic target in PCa, this review aims to provide valuable insights and research directions for clinicians and researchers.
Collapse
Affiliation(s)
- Qiannan Gu
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, Nanjing, Jiangsu 210009, China
| | - Anning Qi
- Medical Laboratory, Liuhe People's Hospital of Jiangsu Province, Nanjing, Jiangsu 211500, China
| | - Ne Wang
- Jiangning Hospital Tiandi New City Branch, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 211198, China
| | - Zhenxian Zhou
- Nanjing Second People's Hospital, Jiangsu Province 211103, China
| | - Xiaohui Zhou
- China Pharmaceutical University, School of Basic Medicine and Clinical Pharmacy, Nanjing, Jiangsu 210009, China; Jiangning Outpatient Department of China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
5
|
Ma S, Ming Y, Wu J, Cui G. Cellular metabolism regulates the differentiation and function of T-cell subsets. Cell Mol Immunol 2024; 21:419-435. [PMID: 38565887 PMCID: PMC11061161 DOI: 10.1038/s41423-024-01148-8] [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: 12/28/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
T cells are an important component of adaptive immunity and protect the host from infectious diseases and cancers. However, uncontrolled T cell immunity may cause autoimmune disorders. In both situations, antigen-specific T cells undergo clonal expansion upon the engagement and activation of antigens. Cellular metabolism is reprogrammed to meet the increase in bioenergetic and biosynthetic demands associated with effector T cell expansion. Metabolites not only serve as building blocks or energy sources to fuel cell growth and expansion but also regulate a broad spectrum of cellular signals that instruct the differentiation of multiple T cell subsets. The realm of immunometabolism research is undergoing swift advancements. Encapsulating all the recent progress within this concise review in not possible. Instead, our objective is to provide a succinct introduction to this swiftly progressing research, concentrating on the metabolic intricacies of three pivotal nutrient classes-lipids, glucose, and amino acids-in T cells. We shed light on recent investigations elucidating the roles of these three groups of metabolites in mediating the metabolic and immune functions of T cells. Moreover, we delve into the prospect of "editing" metabolic pathways within T cells using pharmacological or genetic approaches, with the aim of synergizing this approach with existing immunotherapies and enhancing the efficacy of antitumor and antiinfection immune responses.
Collapse
Affiliation(s)
- Sicong Ma
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Yanan Ming
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Jingxia Wu
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| | - Guoliang Cui
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| |
Collapse
|
6
|
Ewald S, Nasuhidehnavi A, Feng TY, Lesani M, McCall LI. The intersection of host in vivo metabolism and immune responses to infection with kinetoplastid and apicomplexan parasites. Microbiol Mol Biol Rev 2024; 88:e0016422. [PMID: 38299836 PMCID: PMC10966954 DOI: 10.1128/mmbr.00164-22] [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] [Indexed: 02/02/2024] Open
Abstract
SUMMARYProtozoan parasite infection dramatically alters host metabolism, driven by immunological demand and parasite manipulation strategies. Immunometabolic checkpoints are often exploited by kinetoplastid and protozoan parasites to establish chronic infection, which can significantly impair host metabolic homeostasis. The recent growth of tools to analyze metabolism is expanding our understanding of these questions. Here, we review and contrast host metabolic alterations that occur in vivo during infection with Leishmania, trypanosomes, Toxoplasma, Plasmodium, and Cryptosporidium. Although genetically divergent, there are commonalities among these pathogens in terms of metabolic needs, induction of the type I immune responses required for clearance, and the potential for sustained host metabolic dysbiosis. Comparing these pathogens provides an opportunity to explore how transmission strategy, nutritional demand, and host cell and tissue tropism drive similarities and unique aspects in host response and infection outcome and to design new strategies to treat disease.
Collapse
Affiliation(s)
- Sarah Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Azadeh Nasuhidehnavi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
7
|
Meizlish ML, Kimura Y, Pope SD, Matta R, Kim C, Philip NH, Meyaard L, Gonzalez A, Medzhitov R. Mechanosensing regulates tissue repair program in macrophages. SCIENCE ADVANCES 2024; 10:eadk6906. [PMID: 38478620 PMCID: PMC10936955 DOI: 10.1126/sciadv.adk6906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/29/2024] [Indexed: 03/17/2024]
Abstract
Tissue-resident macrophages play important roles in tissue homeostasis and repair. However, how macrophages monitor and maintain tissue integrity is not well understood. The extracellular matrix (ECM) is a key structural and organizational component of all tissues. Here, we find that macrophages sense the mechanical properties of the ECM to regulate a specific tissue repair program. We show that macrophage mechanosensing is mediated by cytoskeletal remodeling and can be performed in three-dimensional environments through a noncanonical, integrin-independent mechanism analogous to amoeboid migration. We find that these cytoskeletal dynamics also integrate biochemical signaling by colony-stimulating factor 1 and ultimately regulate chromatin accessibility to control the mechanosensitive gene expression program. This study identifies an "amoeboid" mode of ECM mechanosensing through which macrophages may regulate tissue repair and fibrosis.
Collapse
Affiliation(s)
- Matthew L. Meizlish
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yoshitaka Kimura
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Scott D. Pope
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Rita Matta
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Catherine Kim
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Naomi H. Philip
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Anjelica Gonzalez
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
8
|
He S, Song W, Cui S, Li J, Jiang Y, Chen X, Peng L. Modulation of miR-146b by N6-methyladenosine modification remodels tumor-associated macrophages and enhances anti-PD-1 therapy in colorectal cancer. Cell Oncol (Dordr) 2023; 46:1731-1746. [PMID: 37402945 DOI: 10.1007/s13402-023-00839-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2023] [Indexed: 07/06/2023] Open
Abstract
PURPOSE MicroRNA-146b (miR-146b) alleviates experimental colitis in mice by mediating macrophage polarization and the release of inflammatory factors. Our goals were to evaluate the antitumor efficacy of miR-146b in colorectal cancer (CRC) and to investigate the underlying mechanisms. METHODS We used murine models of CRC to evaluate whether miR-146b influenced the progression of tumors independent of tumor-associated macrophages (TAMs). RNA immunoprecipitation, N6-methyladenosine (m6A) RNA immunoprecipitation and in vitro pri-miRNA processing assays were conducted to examine whether m6A mediates the maturation of pri-miR-146b/miR-146b. In a series of in vitro and in vivo experiments, we further defined the molecular mechanisms of methyltransferase-like 3 (METTL3)/miR-146b-mediated antitumor immunity and its efficacy in combination with anti-PD-1 immunotherapy. RESULTS We found that miR-146b deletion supported tumor progression by increasing the number of alternatively activated (M2) TAMs. Mechanistically, the m6A-related "writer" protein METTL3 and "reader" protein HNRNPA2B1 controlled miR-146b maturation by regulating the m6A modification region of pri-miR-146b. Furthermore, miR-146b deletion promoted the polarization of M2-TAMs by enhancing phosphoinositide 3-kinase (PI3K)/AKT signaling, and this effect was mediated by the class IA PI3K catalytic subunit p110β, which reduced T cell infiltration, aggravated immunosuppression and ultimately promoted tumor progression. METTL3 knockdown or miR-146b deletion induced programmed death ligand 1 (PD-L1) production via the p110β/PI3K/AKT pathway in TAMs and consequently augmented the antitumor activity of anti-PD-1 immunotherapy. CONCLUSIONS The maturation of pri-miR-146b is m6A-dependent, and miR-146b deletion-mediated TAM differentiation promotes the development of CRC by activating the PI3K/AKT pathway, which induces upregulation of PD-L1 expression, inhibits T cell infiltration into the TME and enhances the antitumor activity of anti-PD-1 immunotherapy. The findings reveal that targeting miR-146b can serve as an adjuvant to anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Shuying He
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 151, Yanjiang West Road, Guangzhou City, 510120, Guangdong Province, China
| | - Wen Song
- Dongguan People's Hospital, Dongguan City, Guangdong Province, China
| | - Shudan Cui
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 151, Yanjiang West Road, Guangzhou City, 510120, Guangdong Province, China
| | - Jiating Li
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 151, Yanjiang West Road, Guangzhou City, 510120, Guangdong Province, China
| | - Yonghong Jiang
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 151, Yanjiang West Road, Guangzhou City, 510120, Guangdong Province, China
| | - Xueqing Chen
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 151, Yanjiang West Road, Guangzhou City, 510120, Guangdong Province, China.
| | - Liang Peng
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 151, Yanjiang West Road, Guangzhou City, 510120, Guangdong Province, China.
| |
Collapse
|
9
|
Brito ASD, Souza JG, Moraes Filho AV, Oliveira PG, Oliveira MAP, Barbosa MS, Sousa JMG, Pinto RM, Conceição EC, Silva CA. Immunostimulating activity of Uncaria tomentosa in RAW 264.7 macrophages. BRAZ J BIOL 2023; 83:e271339. [PMID: 37729210 DOI: 10.1590/1519-6984.271339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/22/2023] [Indexed: 09/22/2023] Open
Abstract
Uncaria tomentosa is a plant native to the Amazon that has immunomodulatory and antitumor properties due to the alkaloids found in the plant, being able to modify the immune response by potentiating or suspending the action of cytokines secreted by macrophages that induce the immune response, either by the classical route (M1) or through the alternative route (M2). Macrophages activated by M1 convert L-arginine into L-citrulline and nitric oxide (NO), whereas macrophages activated by the M2 pathway use the enzymatic activity of arginase to convert the same substrate into L-ornithine and urea. The aim of this work was to evaluate the immunostimulating activity of the crude hydroalcoholic extract from the bark of the U. tomentosa stem in RAW 264.7 macrophages. Concentrations of 0.2, 0.1 and 0.05 mg/mL of U. tomentosa extract associated with LPS, INF-γ and IL-4 inducers were tested by determining NO production and arginase enzyme activity. Nitric oxide production was enhanced by the extract when associated with LPS and LPS + INF-γ inducers. In the activity of the arginase enzyme, the extract decreased the stimulation of IL-4 on the enzyme, mainly at 0.2 mg/mL concentration. Therefore, it is concluded that the crude hydroalcoholic extract of the stem bark of U. tomentosa in RAW 264.7 cells, at a concentration of 0.2 mg/mL, showed considerable pro-inflammatory activity.
Collapse
Affiliation(s)
| | - J G Souza
- Faculdade de Farmácia, Goiânia, GO, Brasil
| | - A V Moraes Filho
- Instituto de Ciências da Saúde, Faculdade Alfredo Nasser, Aparecida de Goiânia, GO, Brasil
| | - P G Oliveira
- Instituto de Patologia Tropical e Saúde Pública, Goiânia, GO, Brasil
| | - M A P Oliveira
- Instituto de Patologia Tropical e Saúde Pública, Goiânia, GO, Brasil
| | - M S Barbosa
- Instituto de Patologia Tropical e Saúde Pública, Goiânia, GO, Brasil
| | - J M G Sousa
- Faculdade de Farmácia, Programa de Pós-graduação em Assistência e Avaliação em Saúde, Goiânia, GO, Brasil
| | - R M Pinto
- Faculdade de Farmácia, Programa de Pós-graduação em Assistência e Avaliação em Saúde, Goiânia, GO, Brasil
| | | | - C A Silva
- Instituto de Patologia Tropical e Saúde Pública, Goiânia, GO, Brasil
| |
Collapse
|
10
|
Han W, Pu H, Li S, Liu Y, Zhao Y, Xu M, Chen C, Wu Y, Yang T, Ye Q, Wang H, Stetler RA, Chen J, Shi Y. Targeted ablation of signal transducer and activator of transduction 1 alleviates inflammation by microglia/macrophages and promotes long-term recovery after ischemic stroke. J Neuroinflammation 2023; 20:178. [PMID: 37516843 PMCID: PMC10385956 DOI: 10.1186/s12974-023-02860-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND Brain microglia and macrophages (Mi/MΦ) can shift to a harmful or advantageous phenotype following an ischemic stroke. Identification of key molecules that regulate the transformation of resting Mi/MΦ could aid in the development of innovative therapies for ischemic stroke. The transcription factor signal transducer and activator of transduction 1 (STAT1) has been found to contribute to acute neuronal death (in the first 24 h) following ischemic stroke, but its effects on Mi/MΦ and influence on long-term stroke outcomes have yet to be determined. METHODS We generated mice with tamoxifen-induced, Mi/MΦ-specific knockout (mKO) of STAT1 driven by Cx3cr1CreER. Expression of STAT1 was examined in the brain by flow cytometry and RNA sequencing after ischemic stroke induced by transient middle cerebral artery occlusion (MCAO). The impact of STAT1 mKO on neuronal cell death, Mi/MΦ phenotype, and brain inflammation profiles were examined 3-5 days after MCAO. Neurological deficits and the integrity of gray and white matter were assessed for 5 weeks after MCAO by various neurobehavioral tests and immunohistochemistry. RESULTS STAT1 was activated in Mi/MΦ at the subacute stage (3 days) after MCAO. Selective deletion of STAT1 in Mi/MΦ did not alter neuronal cell death or infarct size at 24 h after MCAO, but attenuated Mi/MΦ release of high mobility group box 1 and increased arginase 1-producing Mi/MΦ 3d after MCAO, suggesting boosted inflammation-resolving responses of Mi/MΦ. As a result, STAT1 mKO mice had mitigated brain inflammation at the subacute stage after MCAO and less white matter injury in the long term. Importantly, STAT1 mKO was sufficient to improve functional recovery for at least 5 weeks after MCAO in both male and female mice. CONCLUSIONS Mi/MΦ-targeted STAT1 KO does not provide immediate neuroprotection but augments inflammation-resolving actions of Mi/MΦ, thereby facilitating long-term functional recovery after stroke. STAT1 is, therefore, a promising therapeutic target to harness beneficial Mi/MΦ responses and improve long-term outcomes after ischemic stroke.
Collapse
Affiliation(s)
- Wenxuan Han
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Hongjian Pu
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Sicheng Li
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Yaan Liu
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Yongfang Zhao
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Mingyue Xu
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Caixia Chen
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Yun Wu
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Qing Ye
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - R Anne Stetler
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, 3500 Terrace Street, S-510 BST, Pittsburgh, PA, 15213, USA.
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
11
|
Davuluri GVN, Chan CH. Regulation of intrinsic and extrinsic metabolic pathways in tumour-associated macrophages. FEBS J 2023; 290:3040-3058. [PMID: 35486022 PMCID: PMC10711806 DOI: 10.1111/febs.16465] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/08/2022] [Accepted: 04/26/2022] [Indexed: 02/03/2023]
Abstract
Tumour-associated macrophages (TAMs) are highly plastic and are broadly grouped into two major functional states, namely the pro-inflammatory M1-type and the pro-tumoural M2-type. Conversion of the functional states of TAMs is regulated by various cytokines, chemokines growth factors and other secreted factors in the microenvironment. Dysregulated metabolism is a hallmark of cancer. Emerging evidence suggests that metabolism governs the TAM differentiation and functional conversation in support of tumour growth and metastasis. Aside from the altered metabolism reprogramming in TAMs, extracellular metabolites secreted by cancer, stromal and/or other cells within the tumour microenvironment have been found to regulate TAMs through passive competition for metabolite availability and direct regulation via receptor/transporter-mediated signalling reaction. In this review, we focus on the regulatory roles of different metabolites and metabolic pathways in TAM conversion and function. We also discuss if the dysregulated metabolism in TAMs can be exploited for the development of new therapeutic strategies against cancer.
Collapse
Affiliation(s)
| | - Chia-Hsin Chan
- Department of Molecular and Cellular Biology, Roswell Park Cancer Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
12
|
Brigo N, Neumaier E, Pfeifhofer-Obermair C, Grubwieser P, Engl S, Berger S, Seifert M, Reinstadler V, Oberacher H, Weiss G. Timing of Interleukin-4 Stimulation of Macrophages Determines Their Anti-Microbial Activity during Infection with Salmonella enterica Serovar Typhimurium. Cells 2023; 12:1164. [PMID: 37190073 PMCID: PMC10137269 DOI: 10.3390/cells12081164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Priming of macrophages with interferon-gamma (IFNγ) or interleukin-4 (IL-4) leads to polarisation into pro-inflammatory or anti-inflammatory subtypes, which produce key enzymes such as inducible nitric oxide synthase (iNOS) and arginase 1 (ARG1), respectively, and in this way determine host responses to infection. Importantly, L-arginine is the substrate for both enzymes. ARG1 upregulation is associated with increased pathogen load in different infection models. However, while differentiation of macrophages with IL-4 impairs host resistance to the intracellular bacterium Salmonella enterica serovar Typhimurium (S.tm), little is known on the effects of IL-4 on unpolarised macrophages during infection. Therefore, bone-marrow-derived macrophages (BMDM) from C57BL/6N, Tie2Cre+/-ARG1fl/fl (KO), Tie2Cre-/-ARG1fl/fl (WT) mice were infected with S.tm in the undifferentiated state and then stimulated with IL-4 or IFNγ. In addition, BMDM of C57BL/6N mice were first polarised upon stimulation with IL-4 or IFNγ and then infected with S.tm. Interestingly, in contrast to polarisation of BMDM with IL-4 prior to infection, treatment of non-polarised S.tm-infected BMDM with IL-4 resulted in improved infection control whereas stimulation with IFNγ led to an increase in intracellular bacterial numbers compared to unstimulated controls. This effect of IL-4 was paralleled by decreased ARG1 levels and increased iNOS expression. Furthermore, the L-arginine pathway metabolites ornithine and polyamines were enriched in unpolarised cells infected with S.tm and stimulated with IL-4. Depletion of L-arginine reversed the protective effect of IL-4 toward infection control. Our data show that stimulation of S.tm-infected macrophages with IL-4 reduced bacterial multiplication via metabolic re-programming of L-arginine-dependent pathways.
Collapse
Affiliation(s)
- Natascha Brigo
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Emely Neumaier
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Philipp Grubwieser
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Sabine Engl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Sylvia Berger
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Vera Reinstadler
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, Muellerstrasse 44, 6020 Innsbruck, Austria
| | - Herbert Oberacher
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, Muellerstrasse 44, 6020 Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
13
|
Arpa L, Batlle C, Jiang P, Caelles C, Lloberas J, Celada A. Distinct Responses to IL4 in Macrophages Mediated by JNK. Cells 2023; 12:cells12081127. [PMID: 37190036 DOI: 10.3390/cells12081127] [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: 02/10/2023] [Revised: 03/20/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
IL(Interleukin)-4 is the main macrophage M2-type activator and induces an anti-inflammatory phenotype called alternative activation. The IL-4 signaling pathway involves the activation of STAT (Signal Transducer and Activator of Transcription)-6 and members of the MAPK (Mitogen-activated protein kinase) family. In primary-bone-marrow-derived macrophages, we observed a strong activation of JNK (Jun N-terminal kinase)-1 at early time points of IL-4 stimulation. Using selective inhibitors and a knockout model, we explored the contribution of JNK-1 activation to macrophages' response to IL-4. Our findings indicate that JNK-1 regulates the IL-4-mediated expression of genes typically involved in alternative activation, such as Arginase 1 or Mannose receptor, but not others, such as SOCS (suppressor of cytokine signaling) 1 or p21Waf-1 (cyclin dependent kinase inhibitor 1A). Interestingly, we have observed that after macrophages are stimulated with IL-4, JNK-1 has the capacity to phosphorylate STAT-6 on serine but not on tyrosine. Chromatin immunoprecipitation assays revealed that functional JNK-1 is required for the recruitment of co-activators such as CBP (CREB-binding protein)/p300 on the promoter of Arginase 1 but not on p21Waf-1. Taken together, these data demonstrate the critical role of STAT-6 serine phosphorylation by JNK-1 in distinct macrophage responses to IL-4.
Collapse
Affiliation(s)
- Luís Arpa
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Carlos Batlle
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Peijin Jiang
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Carme Caelles
- Institute of Biomedicine, Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Jorge Lloberas
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Antonio Celada
- Biology of Macrophages Group, Department of Cellular Biology, Physiology and Immunology, Universitat de Barcelona, 08007 Barcelona, Spain
| |
Collapse
|
14
|
Mercnik MH, Schliefsteiner C, Fluhr H, Wadsack C. Placental macrophages present distinct polarization pattern and effector functions depending on clinical onset of preeclampsia. Front Immunol 2023; 13:1095879. [PMID: 36713449 PMCID: PMC9878680 DOI: 10.3389/fimmu.2022.1095879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023] Open
Abstract
Hofbauer cells (HBCs) are resident macrophages of the human placenta, regulating immune tolerance and tissue homeostasis. HBCs of a normal placenta (CTR) exhibit mainly an anti-inflammatory M2 phenotype. Under exaggerated chronic inflammation during pregnancy, as in preeclampsia (PE), a phenotypic switch towards M1 polarization has been proposed. PE, defined as maternally derived syndrome can be distinguished into two different entities: early-onset (EO) preeclampsia and late-onset (LO) preeclampsia. Although the clinical presenting characteristics overlap, both can be identified by biochemical markers, heritability, and different maternal and fetal outcomes. To date, no study has specifically investigated polarization and phenotype of EO- and LO-PE HBCs and looked at possible changes in HBC functionality. Primary HBCs were isolated from CTR and PE placentae. First, in vitro morphological differences were observed between CTR and PE HBCs, with both PE groups exhibiting features of M1 macrophages alongside M2 forms. Interestingly, a different polarization pattern was observed between EO- and LO-PE HBCs. EO-PE HBCs develop a tissue remodeling M2 phenotype that is strongly shifted toward M1 polarization and showed a significant upregulation of CD86, TLR4, and HLA-DR. Furthermore, this pro-inflammatory signature is corroborated by higher expression of IRF5 and of NOS2 (p ≤ 0.05). However, their M2 characteristics is reflected by significant TGF-β secretion and ARG1 expression. In contrast, LO-PE HBCs developed a phagocytic CD209-low M2 phenotype in which the M1 pattern was not as pronounced as they downregulated the NOS2 gene, but expressed increased levels of pro-inflammatory CD80 and TLR1 (p ≤ 0.05). The enhanced phagocytosis and MMP-9 secretion alongside the increased secretion of anti-inflammatory IL -4, IL -13 and TGF-β in both EO- and LO-PE HBCs suggests their adaptive role and plasticity in resolving inflammation and tissue homeostasis.
Collapse
Affiliation(s)
| | | | - Herbert Fluhr
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria,BioTechMed-Graz, Graz, Austria,*Correspondence: Christian Wadsack,
| |
Collapse
|
15
|
Li X, Gulati M, Larson AC, Solheim JC, Jain M, Kumar S, Batra SK. Immune checkpoint blockade in pancreatic cancer: Trudging through the immune desert. Semin Cancer Biol 2022; 86:14-27. [PMID: 36041672 PMCID: PMC9713834 DOI: 10.1016/j.semcancer.2022.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/01/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022]
Abstract
Pancreatic cancer (PC) has exceptionally high mortality due to ineffective treatment strategies. Immunotherapy, which mobilizes the immune system to fight against cancer, has been proven successful in multiple cancers; however, its application in PC has met with limited success. In this review, we articulated that the pancreatic tumor microenvironment is immuno-suppressive with extensive infiltration by M2-macrophages and myeloid-derived suppressive cells but low numbers of cytotoxic T-cells. In addition, low mutational load and poor antigen processing, presentation, and recognition contribute to the limited response to immunotherapy in PC. Immune checkpoints, the critical targets for immunotherapy, have high expression in PC and stromal cells, regulated by tumor microenvironmental milieu (cytokine and metabolites) and cell-intrinsic mechanisms (epigenetic regulation, oncogenic signaling, and post-translational modifications). Combining immunotherapy with modulators of the tumor microenvironment may facilitate the development of novel therapeutic regimens to manage PC.
Collapse
Affiliation(s)
- Xiaoqi Li
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mansi Gulati
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Alaina C Larson
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joyce C Solheim
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
16
|
Wang J, Long R, Han Y. The role of exosomes in the tumour microenvironment on macrophage polarisation. Biochim Biophys Acta Rev Cancer 2022; 1877:188811. [DOI: 10.1016/j.bbcan.2022.188811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/15/2022] [Accepted: 09/28/2022] [Indexed: 12/14/2022]
|
17
|
Crosstalk between PI3K/AKT/KLF4 signaling and microglia M1/M2 polarization as a novel mechanistic approach towards flibanserin repositioning in parkinson's disease. Int Immunopharmacol 2022; 112:109191. [PMID: 36055034 DOI: 10.1016/j.intimp.2022.109191] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 02/07/2023]
Abstract
Balancing microglia M1/M2 polarization has been shown as a prospective therapeutic strategy for Parkinson's disease (PD). Various vital signaling pathways are likely to govern the microglial phenotype. The implication of 5HT1A receptors in neurodegenerative disorders has raised interest in exploring the repositioning of flibanserin (Flib), a 5HT1A agonist, as an effective neuroprotective agent for PD. Therefore, this study was designed to assess the ability of Flib to modulate microglia phenotype switching from M1 to M2 via PI3K/AKT downstream targets in a rotenone model of PD. Rats received rotenone (1.5 mg/kg) every other day and were concurrently treated with Flib (40 mg/kg/day) with or without wortmannin (15 μg/kg/day), a PI3K inhibitor, for 21 days. Flib improved the motor perturbations induced by rotenone, as confirmed by the reversion of histopathological damage and tyrosine hydroxylase immunohistochemical alterations in both the striata and substantia nigra. The molecular signaling of Flib was elaborated by inducing striatal AKT phosphorylation and the expression of its substantial target, KLF4. Flib induced STAT6 phosphorylation to promote M2 polarization as demonstrated by the increased CD163++ microglial count with striatal arginase activity. In parallel, it markedly inhibited M1 activation as evidenced by the reduction in CD86++ microglia count with striatal proinflammatory mediators, IL-1β and iNOS. The pre-administration of wortmannin mostly negated Flib's neuroprotective effects. In conclusion, Flib AKT/ KLF4-dependently amended M1/M2 microglial imbalance to exert a promising neuroprotective effect, highlighting its potential as a revolutionary candidate for conquering PD.
Collapse
|
18
|
Opioid receptor activation suppresses the neuroinflammatory response by promoting microglial M2 polarization. Mol Cell Neurosci 2022; 121:103744. [PMID: 35660086 DOI: 10.1016/j.mcn.2022.103744] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/02/2022] [Accepted: 05/29/2022] [Indexed: 11/20/2022] Open
Abstract
Activation of microglia is considered the most important component of neuroinflammation. Microglia can adopt a pro-inflammatory (M1) or anti-inflammatory (M2) phenotype. Opioid receptors (ORs) have been shown to control neurotransmission of various peptidergic neurons, but their potential role in regulating microglial function is largely unknown. Here, we aimed to investigate the effect of the OR agonists DAMGO, DADLE and U-50488 on the polarization of C8-B4 microglial cells. We observed that opioids suppressed lipopolysaccharide (LPS)-triggered M1 polarization and promoted M2 polarization. This was reflected in lower phagocytic activity, lower production of NO, lower expression of TNF-α, IL-1β, IL-6, IL-86 and IL-12 beta p40 together with higher migration rate, and increased expression of IL-4, IL-10, arginase 1 and CD 206 in microglia, compared to cells affected by LPS. We demonstrated that the effect of opioids on microglial polarization is mediated by the TREM2/NF-κB signaling pathway. These results provide new insights into the anti-inflammatory and neuroprotective effects of opioids and highlight their potential in combating neurodegenerative diseases.
Collapse
|
19
|
Sheida F, Razi S, Keshavarz-Fathi M, Rezaei N. The role of myeloid-derived suppressor cells in lung cancer and targeted immunotherapies. Expert Rev Anticancer Ther 2021; 22:65-81. [PMID: 34821533 DOI: 10.1080/14737140.2022.2011224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Lung cancer is the deadliest cancer in both sexes combined globally due to significant delays in diagnosis and poor survival. Despite advances in the treatment of lung cancer, the overall outcomes remain poor and traditional chemotherapy fails to provide long-term benefits for many patients. Therefore, new treatment strategies are needed to increase overall survival. Myeloid-derived suppressor cells (MDSCs) are immunosuppressive cells taking part in lung cancer, as has been described in other types of tumors. MDSCs immunosuppressive activity is mediated by arginases (ARG-1 and ARG-2), nitric oxide (NO), reactive oxygen species (ROS), peroxynitrite, PD-1/PD-L1 axis, and different cytokines. MDSCs can be a target for lung cancer immunotherapy by inducing their differentiation into mature myeloid cells, elimination, attenuation of their function, and inhibition of their accumulation. AREAS COVERED In this review, the immunosuppressive function of MDSCs, their role in lung cancer, and strategies to target them, which could result in increased efficacy of immunotherapy in patients with lung cancer, are discussed. EXPERT OPINION Identification of important mechanisms and upstream pathways involved in MDSCs functions paves the way for further preclinical and clinical lung cancer research, which could lead to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Fateme Sheida
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 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.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 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), Stockholm, Sweden
| |
Collapse
|
20
|
Gao ZW, Li L, Huang YY, Zhao CQ, Xue SJ, Chen J, Yang ZZ, Xu JF, Su X. Vagal-α7nAChR signaling is required for lung anti-inflammatory responses and arginase 1 expression during an influenza infection. Acta Pharmacol Sin 2021; 42:1642-1652. [PMID: 33414508 PMCID: PMC8463540 DOI: 10.1038/s41401-020-00579-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/16/2020] [Indexed: 02/02/2023] Open
Abstract
Vagal circuit-α7 nicotinic acetylcholine receptor (α7nAChR, coded by Chrna7) signaling can modulate lung proinflammatory responses. Arginase 1 (ARG1) plays a crucial role in the resolution of lung inflammation. However, whether vagal-α7nAChR signaling can regulate lung inflammation and ARG1 expression during an influenza infection is elusive. Here, we found that lung and spleen IL-4+ cells and lung ARG1 expression were reduced; however, bronchoalveolar lavage (BAL) protein and leukocytes and lung inflammatory cytokines were increased in PR8 (A/Puerto Rico/8/1934, H1N1)-infected vagotomized mice when compared to the control. In PR8-infected α7nAChR-deficient mice, lung Arg1, Il10, and Socs3 expression and BAL Ly6C+CD206+ cells were reduced. PR8-infected Chrna7+/+ recipient mice reconstituted with Chrna7-/- bone marrow had a lower survival as compared to PR8-infected Chrna7+/+ recipient mice reconstituted with Chrna7+/+ bone marrow. Mechanistically, the activation of α7nAChR by its agonist GTS-21 could enhance IL-4-induced Arg1 expression, reduced Nos2, and TNF-α expression in PR8-infected bone marrow-derived macrophages (BMDM). Stimulation with IL-4 increased phosphorylation of STAT6 and activation of α7nAChR increased STAT6 binding with the ARG1 promoter and relieved IL-4-induced H3K27me3 methylation by increasing JMJD3 expression in PR8-infected BMDM. Inhibition of JMJD3 increased H3K27me3 methylation and abolished α7nAChR activation and IL-4 induced ARG1 expression. Activation of α7nAChR also reduced phosphorylation of AKT1 and contained FOXO1 in the nucleus. Knockdown of Foxo1a reduced α7nAChR activation and IL-4 induced Arg1 expression in PR8-infected BMDM. Therefore, vagal-α7nAChR signaling is a novel therapeutic target for treating lung inflammatory responses during an influenza infection.
Collapse
Affiliation(s)
- Zhao-Wei Gao
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ling Li
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuan-Yuan Huang
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Cai-Qi Zhao
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shuang-Jia Xue
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jie Chen
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhong-Zhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Jin-Fu Xu
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, 200433, China.
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
21
|
Fu W, Hu W, Yi YS, Hettinghouse A, Sun G, Bi Y, He W, Zhang L, Gao G, Liu J, Toyo-Oka K, Xiao G, Solit DB, Loke P, Liu CJ. TNFR2/14-3-3ε signaling complex instructs macrophage plasticity in inflammation and autoimmunity. J Clin Invest 2021; 131:e144016. [PMID: 34185706 DOI: 10.1172/jci144016] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 06/25/2021] [Indexed: 12/13/2022] Open
Abstract
TNFR1 and TNFR2 have received prominent attention because of their dominance in the pathogenesis of inflammation and autoimmunity. TNFR1 has been extensively studied and primarily mediates inflammation. TNFR2 remains far less studied, although emerging evidence demonstrates that TNFR2 plays an antiinflammatory and immunoregulatory role in various conditions and diseases. Herein, we report that TNFR2 regulates macrophage polarization, a highly dynamic process controlled by largely unidentified intracellular regulators. Using biochemical copurification and mass spectrometry approaches, we isolated the signaling molecule 14-3-3ε as a component of TNFR2 complexes in response to progranulin stimulation in macrophages. In addition, 14-3-3ε was essential for TNFR2 signaling-mediated regulation of macrophage polarization and switch. Both global and myeloid-specific deletion of 14-3-3ε resulted in exacerbated inflammatory arthritis and counteracted the protective effects of progranulin-mediated TNFR2 activation against inflammation and autoimmunity. TNFR2/14-3-3ε signaled through PI3K/Akt/mTOR to restrict NF-κB activation while simultaneously stimulating C/EBPβ activation, thereby instructing macrophage plasticity. Collectively, this study identifies 14-3-3ε as a previously unrecognized vital component of the TNFR2 receptor complex and provides new insights into the TNFR2 signaling, particularly its role in macrophage polarization with therapeutic implications for various inflammatory and autoimmune diseases with activation of the TNFR2/14-3-3ε antiinflammatory pathway.
Collapse
Affiliation(s)
- Wenyu Fu
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Wenhuo Hu
- Human Oncology and Pathogenesis Program and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Young-Su Yi
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Aubryanna Hettinghouse
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Guodong Sun
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Yufei Bi
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Wenjun He
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Lei Zhang
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Guanmin Gao
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Jody Liu
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China
| | - David B Solit
- Human Oncology and Pathogenesis Program and Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Png Loke
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Chuan-Ju Liu
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, New York, USA.,Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
22
|
Martí I Líndez AA, Reith W. Arginine-dependent immune responses. Cell Mol Life Sci 2021; 78:5303-5324. [PMID: 34037806 PMCID: PMC8257534 DOI: 10.1007/s00018-021-03828-4] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
A growing body of evidence indicates that, over the course of evolution of the immune system, arginine has been selected as a node for the regulation of immune responses. An appropriate supply of arginine has long been associated with the improvement of immune responses. In addition to being a building block for protein synthesis, arginine serves as a substrate for distinct metabolic pathways that profoundly affect immune cell biology; especially macrophage, dendritic cell and T cell immunobiology. Arginine availability, synthesis, and catabolism are highly interrelated aspects of immune responses and their fine-tuning can dictate divergent pro-inflammatory or anti-inflammatory immune outcomes. Here, we review the organismal pathways of arginine metabolism in humans and rodents, as essential modulators of the availability of this semi-essential amino acid for immune cells. We subsequently review well-established and novel findings on the functional impact of arginine biosynthetic and catabolic pathways on the main immune cell lineages. Finally, as arginine has emerged as a molecule impacting on a plethora of immune functions, we integrate key notions on how the disruption or perversion of arginine metabolism is implicated in pathologies ranging from infectious diseases to autoimmunity and cancer.
Collapse
Affiliation(s)
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
23
|
Juban G. Transcriptional control of macrophage inflammatory shift during skeletal muscle regeneration. Semin Cell Dev Biol 2021; 119:82-88. [PMID: 34183241 DOI: 10.1016/j.semcdb.2021.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/13/2021] [Accepted: 06/18/2021] [Indexed: 01/05/2023]
Abstract
Skeletal muscle is a tissue able to fully regenerate after an acute injury. Macrophages play an essential role during skeletal muscle regeneration. Resolution of inflammation is a crucial step during the regeneration process, allowing to contain the inflammatory response to avoid damage of the healthy surrounding muscle and triggers the recovery phase during which the muscle regenerates. Resolution of inflammation is mainly mediated by macrophage phenotypic shift that is the transition from a pro-inflammatory damage associated profile towards an anti-inflammatory restorative phenotype, which is characterized by a major transcriptional rewiring. Failure of the resolution of inflammation is observed in chronic diseases such as degenerative myopathies where permanent asynchronous muscle injuries trigger contradictory inflammatory cues, leading to fibrosis and alteration of muscle function. This review will focus on the described molecular pathways that control macrophage inflammatory shift during skeletal muscle regeneration. First, we will highlight the transcriptional changes that characterize macrophage inflammatory shift during skeletal muscle regeneration. Then, we will describe how the signaling pathways and the metabolic changes associated with this shift are controlled. Finally, we will emphasize the transcription factors involved.
Collapse
Affiliation(s)
- Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, Lyon, France.
| |
Collapse
|
24
|
Amino acid metabolism and signalling pathways: potential targets in the control of infection and immunity. Eur J Clin Nutr 2021; 75:1319-1327. [PMID: 34163018 PMCID: PMC8220356 DOI: 10.1038/s41430-021-00943-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/13/2022]
Abstract
Defences to pathogens such as SarCoV2 in mammals involves interactions between immune functions and metabolic pathways to eradicate infection while preventing hyperinflammation. Amino acid metabolic pathways represent with other antimicrobial agent potential targets for therapeutic strategies. iNOS-mediated production of NO from Arg is involved in the innate inflammatory response to pathogens and NO overproduction can induce hyperinflammation. The two Arg- and Trp-catabolising enzymes Arg1 and IDO1 reduce the hyperinflammation by an immunosuppressive effect via either Arg starvation (for Arg1) or via the immunoregulatory activity of the Trp-derived metabolites Kyn (for IDO1). In response to amino acid abundance mTOR activates the host protein translation and Coronaviruses use this machinery for their own protein synthesis and replication. In contrast GCN2, the sensor of amino acid starvation, activates pathways that restrict inflammation and viral replication. Gln depletion alters the immune response that become more suppressive, by favouring a regulatory T phenotype rather than a Th1 phenotype. Proliferating activated immune cells are highly dependent on Ser, activation and differentiation of T cells need enough Ser and dietary Ser restriction can inhibit their proliferation. Cys is strictly required for T-cell proliferation because they cannot convert Met to Cys. Restricting Met inhibits both viral RNA cap methylation and replication, and the proliferation of infected cells with an increased requirement for Met. Phe catabolism produces antimicrobial metabolites resulting in the inhibition of microbial growth and an immunosuppressive activity towards T lymphocytes.
Collapse
|
25
|
Tomé D. Amino acid metabolism and signalling pathways: potential targets in the control of infection and immunity. Nutr Diabetes 2021; 11:20. [PMID: 34168115 PMCID: PMC8223530 DOI: 10.1038/s41387-021-00164-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
Defences to pathogens such as SarCoV2 in mammals involves interactions between immune functions and metabolic pathways to eradicate infection while preventing hyperinflammation. Amino acid metabolic pathways represent with other antimicrobial agent potential targets for therapeutic strategies. iNOS-mediated production of NO from Arg is involved in the innate inflammatory response to pathogens and NO overproduction can induce hyperinflammation. The two Arg-catabolising enzymes Arg1 and IDO1 reduce the hyperinflammation by an immunosuppressive effect via either Arg starvation (for Arg1) or via the immunoregulatory activity of the Arg-derived metabolites Kyn (for IDO1). In response to amino acid abundance mTOR activates the host protein translation and Coronaviruses use this machinery for their own protein synthesis and replication. In contrast GCN2, the sensor of amino acid starvation, activates pathways that restrict inflammation and viral replication. Gln depletion alters the immune response that become more suppressive, by favouring a regulatory T phenotype rather than a Th1 phenotype. Proliferating activated immune cells are highly dependent on Ser, activation and differentiation of T cells need enough Ser and dietary Ser restriction can inhibit their proliferation. Cys is strictly required for T-cell proliferation because they cannot convert Met to Cys. Restricting Met inhibits both viral RNA cap methylation and replication, and the proliferation of infected cells with an increased requirement for Met. Phe catabolism produces antimicrobial metabolites resulting in the inhibition of microbial growth and an immunosuppressive activity towards T lymphocytes.
Collapse
Affiliation(s)
- Daniel Tomé
- grid.417885.70000 0001 2185 8223UMR PNCA, AgroParisTech, INRAE, Université Paris-Saclay, Paris, France
| |
Collapse
|
26
|
Mike JK, Ferriero DM. Efferocytosis Mediated Modulation of Injury after Neonatal Brain Hypoxia-Ischemia. Cells 2021; 10:1025. [PMID: 33925299 PMCID: PMC8146813 DOI: 10.3390/cells10051025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Neonatal brain hypoxia-ischemia (HI) is a leading cause of morbidity and long-term disabilities in children. While we have made significant progress in describing HI mechanisms, the limited therapies currently offered for HI treatment in the clinical setting stress the importance of discovering new targetable pathways. Efferocytosis is an immunoregulatory and homeostatic process of clearance of apoptotic cells (AC) and cellular debris, best described in the brain during neurodevelopment. The therapeutic potential of stimulating defective efferocytosis has been recognized in neurodegenerative diseases. In this review, we will explore the involvement of efferocytosis after a stroke and HI as a promising target for new HI therapies.
Collapse
Affiliation(s)
- Jana Krystofova Mike
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Donna Marie Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
- Department of Neurology Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
27
|
Liu W, Taso O, Wang R, Bayram S, Graham AC, Garcia-Reitboeck P, Mallach A, Andrews WD, Piers TM, Botia JA, Pocock JM, Cummings DM, Hardy J, Edwards FA, Salih DA. Trem2 promotes anti-inflammatory responses in microglia and is suppressed under pro-inflammatory conditions. Hum Mol Genet 2020; 29:3224-3248. [PMID: 32959884 PMCID: PMC7689298 DOI: 10.1093/hmg/ddaa209] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022] Open
Abstract
Genome-wide association studies have reported that, amongst other microglial genes, variants in TREM2 can profoundly increase the incidence of developing Alzheimer's disease (AD). We have investigated the role of TREM2 in primary microglial cultures from wild type mice by using siRNA to decrease Trem2 expression, and in parallel from knock-in mice heterozygous or homozygous for the Trem2 R47H AD risk variant. The prevailing phenotype of Trem2 R47H knock-in mice was decreased expression levels of Trem2 in microglia, which resulted in decreased density of microglia in the hippocampus. Overall, primary microglia with reduced Trem2 expression, either by siRNA or from the R47H knock-in mice, displayed a similar phenotype. Comparison of the effects of decreased Trem2 expression under conditions of lipopolysaccharide (LPS) pro-inflammatory or IL-4 anti-inflammatory stimulation revealed the importance of Trem2 in driving a number of the genes up-regulated in the anti-inflammatory phenotype. RNA-seq analysis showed that IL-4 induced the expression of a program of genes including Arg1 and Ap1b1 in microglia, which showed an attenuated response to IL-4 when Trem2 expression was decreased. Genes showing a similar expression profile to Arg1 were enriched for STAT6 transcription factor recognition elements in their promoter, and Trem2 knockdown decreased levels of STAT6. LPS-induced pro-inflammatory stimulation suppressed Trem2 expression, thus preventing TREM2's anti-inflammatory drive. Given that anti-inflammatory signaling is associated with tissue repair, understanding the signaling mechanisms downstream of Trem2 in coordinating the pro- and anti-inflammatory balance of microglia, particularly mediating effects of the IL-4-regulated anti-inflammatory pathway, has important implications for fighting neurodegenerative disease.
Collapse
Affiliation(s)
- Wenfei Liu
- Department of Neuroscience, Physiology and Pharmacology, UCL, London WC1E 6BT, UK
| | - Orjona Taso
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Rui Wang
- Department of Neuroscience, Physiology and Pharmacology, UCL, London WC1E 6BT, UK
| | | | | | | | - Anna Mallach
- Department of Neuroinflammation, Institute of Neurology, UCL, London WC1N 1PJ, UK
| | - William D Andrews
- Department of Cell and Developmental Biology, UCL, London WC1E 6BT, UK
| | - Thomas M Piers
- Department of Neuroinflammation, Institute of Neurology, UCL, London WC1N 1PJ, UK
| | - Juan A Botia
- Department of Information and Communications Engineering, Universidad de Murcia, Murcia E-30100, Spain
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, London WC1N 1PJ, UK
| | - Jennifer M Pocock
- Department of Neuroinflammation, Institute of Neurology, UCL, London WC1N 1PJ, UK
| | - Damian M Cummings
- Department of Neuroscience, Physiology and Pharmacology, UCL, London WC1E 6BT, UK
| | - John Hardy
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Department of Neurodegenerative Diseases, Institute of Neurology, UCL, London WC1N 1PJ, UK
| | - Frances A Edwards
- Department of Neuroscience, Physiology and Pharmacology, UCL, London WC1E 6BT, UK
| | - Dervis A Salih
- Department of Neuroscience, Physiology and Pharmacology, UCL, London WC1E 6BT, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| |
Collapse
|
28
|
Weng X, Zhao H, Guan Q, Shi G, Feng S, Gleave ME, Nguan CC, Du C. Clusterin regulates macrophage expansion, polarization and phagocytic activity in response to inflammation in the kidneys. Immunol Cell Biol 2020; 99:274-287. [PMID: 32935392 PMCID: PMC7984284 DOI: 10.1111/imcb.12405] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/22/2020] [Accepted: 09/13/2020] [Indexed: 12/26/2022]
Abstract
Clusterin (CLU) is a multifunctional protein localized extracellularly and intracellularly. Although CLU-knockout (KO) mice are more susceptible to renal ischemia-reperfusion injury (IRI), the mechanisms underlying the actions of CLU in IRI are not fully understood. Macrophages are key regulators of IRI severity and tissue repair. Therefore, we investigated the role of CLU in macrophage polarization and phagocytosis. Renal IRI was induced in wild-type (WT) or CLU-KO C57BL/6 mice by clamping the renal pedicles for 30 min at 32°C. Peritoneal macrophages were activated via an intraperitoneal injection of lipopolysaccharide (LPS). Renal tissue damage was examined using histology, whereas leukocyte phenotypes were assessed using flow cytometry and immunohistochemistry. We found that monocytes/macrophages expressed the CLU protein that was upregulated by hypoxia. The percentages of macrophages (F4/80+ , CD11b+ or MAC3+ ) infiltrating the kidneys of WT mice were significantly less than those in CLU-KO mice after IRI. The M1/M2 phenotype ratio of the macrophages in WT kidneys decreased at day 7 post-IRI when the injury was repaired, whereas that in KO kidneys increased consistently as tissue injury persisted. In response to LPS stimulation, WT mice produced fewer M1 macrophages, but not M2, than the control did. Phagocytosis was stimulated by CLU expression in macrophages compared with the CLU null controls and by the exogenous CLU protein. In conclusion, CLU suppresses macrophage infiltration and proinflammatory M1 polarization during the recovery period following IRI, and enhances phagocytic activity, which may be partly responsible for tissue repair in the kidneys of WT mice after injury.
Collapse
Affiliation(s)
- Xiaodong Weng
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.,Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Haimei Zhao
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.,College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Qiunong Guan
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Ganggang Shi
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.,Department of Colorectal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Shijian Feng
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Martin E Gleave
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Christopher Cy Nguan
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Caigan Du
- Department of Urologic Sciences, The University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| |
Collapse
|
29
|
Luiz JPM, Toller‐Kawahisa JE, Viacava PR, Nascimento DC, Pereira PT, Saraiva AL, Prado DS, Le Bert M, Giurisato E, Tournier C, Cunha TM, Cunha FQ, Quesniaux V, Ryffel B, Alves‐Filho JC. MEK5/ERK5 signaling mediates IL‐4‐induced M2 macrophage differentiation through regulation of c‐Myc expression. J Leukoc Biol 2020; 108:1215-1223. [DOI: 10.1002/jlb.1ma0520-016r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/27/2020] [Accepted: 07/16/2020] [Indexed: 01/10/2023] Open
Affiliation(s)
- João Paulo M. Luiz
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Juliana E. Toller‐Kawahisa
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Paula R. Viacava
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Daniele C. Nascimento
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Priscilla T. Pereira
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - André L. Saraiva
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Douglas S. Prado
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Marc Le Bert
- CNRS, UMR7355 Orléans, France
- Experimental and Molecular Immunology and Neurogenetics University of Orléans Orléans France
| | - Emanuele Giurisato
- Division of Cancer Sciences School of Medical Sciences Faculty of Biology, Medicine and Health University of Manchester Manchester United Kingdom
| | - Cathy Tournier
- Division of Cancer Sciences School of Medical Sciences Faculty of Biology, Medicine and Health University of Manchester Manchester United Kingdom
| | - Thiago M. Cunha
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Fernando Q. Cunha
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Valerie Quesniaux
- CNRS, UMR7355 Orléans, France
- Experimental and Molecular Immunology and Neurogenetics University of Orléans Orléans France
| | - Bernhard Ryffel
- CNRS, UMR7355 Orléans, France
- Experimental and Molecular Immunology and Neurogenetics University of Orléans Orléans France
| | - José C. Alves‐Filho
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| |
Collapse
|
30
|
Tibbo AJ, Baillie GS. Phosphodiesterase 4B: Master Regulator of Brain Signaling. Cells 2020; 9:cells9051254. [PMID: 32438615 PMCID: PMC7291338 DOI: 10.3390/cells9051254] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022] Open
Abstract
Phosphodiesterases (PDEs) are the only superfamily of enzymes that have the ability to break down cyclic nucleotides and, as such, they have a pivotal role in neurological disease and brain development. PDEs have a modular structure that allows targeting of individual isoforms to discrete brain locations and it is often the location of a PDE that shapes its cellular function. Many of the eleven different families of PDEs have been associated with specific diseases. However, we evaluate the evidence, which suggests the activity from a sub-family of the PDE4 family, namely PDE4B, underpins a range of important functions in the brain that positions the PDE4B enzymes as a therapeutic target for a diverse collection of indications, such as, schizophrenia, neuroinflammation, and cognitive function.
Collapse
|
31
|
Grzywa TM, Sosnowska A, Matryba P, Rydzynska Z, Jasinski M, Nowis D, Golab J. Myeloid Cell-Derived Arginase in Cancer Immune Response. Front Immunol 2020; 11:938. [PMID: 32499785 PMCID: PMC7242730 DOI: 10.3389/fimmu.2020.00938] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Amino acid metabolism is a critical regulator of the immune response, and its modulating becomes a promising approach in various forms of immunotherapy. Insufficient concentrations of essential amino acids restrict T-cells activation and proliferation. However, only arginases, that degrade L-arginine, as well as enzymes that hydrolyze L-tryptophan are substantially increased in cancer. Two arginase isoforms, ARG1 and ARG2, have been found to be present in tumors and their increased activity usually correlates with more advanced disease and worse clinical prognosis. Nearly all types of myeloid cells were reported to produce arginases and the increased numbers of various populations of myeloid-derived suppressor cells and macrophages correlate with inferior clinical outcomes of cancer patients. Here, we describe the role of arginases produced by myeloid cells in regulating various populations of immune cells, discuss molecular mechanisms of immunoregulatory processes involving L-arginine metabolism and outline therapeutic approaches to mitigate the negative effects of arginases on antitumor immune response. Development of potent arginase inhibitors, with improved pharmacokinetic properties, may lead to the elaboration of novel therapeutic strategies based on targeting immunoregulatory pathways controlled by L-arginine degradation.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Anna Sosnowska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Paweł Matryba
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Neurobiology BRAINCITY, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
- The Doctoral School of the Medical University of Warsaw, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Rydzynska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Jasinski
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Experimental Medicine, Center of New Technologies, University of Warsaw, Warsaw, Poland
- Genomic Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Centre of Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
32
|
Canè S, Bronte V. Detection and functional evaluation of arginase-1 isolated from human PMNs and murine MDSC. Methods Enzymol 2020; 632:193-213. [DOI: 10.1016/bs.mie.2019.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Zhao C, Mirando AC, Sové RJ, Medeiros TX, Annex BH, Popel AS. A mechanistic integrative computational model of macrophage polarization: Implications in human pathophysiology. PLoS Comput Biol 2019; 15:e1007468. [PMID: 31738746 PMCID: PMC6860420 DOI: 10.1371/journal.pcbi.1007468] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022] Open
Abstract
Macrophages respond to signals in the microenvironment by changing their functional phenotypes, a process known as polarization. Depending on the context, they acquire different patterns of transcriptional activation, cytokine expression and cellular metabolism which collectively constitute a continuous spectrum of phenotypes, of which the two extremes are denoted as classical (M1) and alternative (M2) activation. To quantitatively decode the underlying principles governing macrophage phenotypic polarization and thereby harness its therapeutic potential in human diseases, a systems-level approach is needed given the multitude of signaling pathways and intracellular regulation involved. Here we develop the first mechanism-based, multi-pathway computational model that describes the integrated signal transduction and macrophage programming under M1 (IFN-γ), M2 (IL-4) and cell stress (hypoxia) stimulation. Our model was calibrated extensively against experimental data, and we mechanistically elucidated several signature feedbacks behind the M1-M2 antagonism and investigated the dynamical shaping of macrophage phenotypes within the M1-M2 spectrum. Model sensitivity analysis also revealed key molecular nodes and interactions as targets with potential therapeutic values for the pathophysiology of peripheral arterial disease and cancer. Through simulations that dynamically capture the signal integration and phenotypic marker expression in the differential macrophage polarization responses, our model provides an important computational basis toward a more quantitative and network-centric understanding of the complex physiology and versatile functions of macrophages in human diseases.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| | - Adam C. Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Richard J. Sové
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Thalyta X. Medeiros
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States of America
- Divison of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Brian H. Annex
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States of America
- Divison of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
34
|
Lamkin DM, Srivastava S, Bradshaw KP, Betz JE, Muy KB, Wiese AM, Yee SK, Waggoner RM, Arevalo JMG, Yoon AJ, Faull KF, Sloan EK, Cole SW. C/EBPβ regulates the M2 transcriptome in β-adrenergic-stimulated macrophages. Brain Behav Immun 2019; 80:839-848. [PMID: 31132458 PMCID: PMC6660400 DOI: 10.1016/j.bbi.2019.05.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/11/2022] Open
Abstract
At the M2 terminal of the macrophage activation spectrum, expression of genes is regulated by transcription factors that include STAT6, CREB, and C/EBPβ. Signaling through β-adrenergic receptors drives M2 activation of macrophages, but little is known about the transcription factors involved. In the present study, we found that C/EBPβ regulates the signaling pathway between β-adrenergic stimulation and expression of Arg1 and several other specific genes in the greater M2 transcriptome. β-adrenergic signaling induced Cebpb gene expression relatively early with a peak at 1 h post-stimulation, followed by peak Arg1 gene expression at 8 h. C/EBPβ transcription factor activity was elevated at the enhancer region for Arg 1 at both 4 and 8 h after stimulation but not near the more proximal promoter region. Knockdown of Cebpb suppressed the β-adrenergic-induced peak in Cebpb gene expression as well as subsequent accumulation of C/EBPβ protein in the nucleus, which resulted in suppression of β-adrenergic-induced Arg1 gene expression. Analysis of genome-wide transcriptional profiles identified 20 additional M2 genes that followed the same pattern of regulation by β-adrenergic- and C/EBPβ-signaling. Promoter-based bioinformatic analysis confirmed enrichment of binding motifs for C/EBPβ transcription factor across these M2 genes. These findings pinpoint a mechanism that may be targeted to redirect the deleterious influence of β-adrenergic signaling on macrophage involvement in M2-related diseases such as cancer.
Collapse
Affiliation(s)
- Donald M Lamkin
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States; Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles 90095, United States; Jonsson Comprehensive Cancer Center, University of California, Los Angeles 90095, United States.
| | - Shreyesi Srivastava
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Karen P Bradshaw
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Jenna E Betz
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Kevin B Muy
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Anna M Wiese
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Shelby K Yee
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Rebecca M Waggoner
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Jesusa M G Arevalo
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States; Divison of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles 90095, United States
| | - Alexander J Yoon
- Pasarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Kym F Faull
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles 90095, United States; Pasarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States
| | - Erica K Sloan
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States; Jonsson Comprehensive Cancer Center, University of California, Los Angeles 90095, United States; Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, VIC 3002, Australia
| | - Steve W Cole
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles 90095, United States; Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles 90095, United States; Jonsson Comprehensive Cancer Center, University of California, Los Angeles 90095, United States; Divison of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles 90095, United States
| |
Collapse
|
35
|
Heung LJ, Hohl TM. Inflammatory monocytes are detrimental to the host immune response during acute infection with Cryptococcus neoformans. PLoS Pathog 2019; 15:e1007627. [PMID: 30897162 PMCID: PMC6428256 DOI: 10.1371/journal.ppat.1007627] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/07/2019] [Indexed: 12/30/2022] Open
Abstract
Cryptococcus neoformans is a leading cause of invasive fungal infections among immunocompromised patients. However, the cellular constituents of the innate immune response that promote clearance versus progression of infection upon respiratory acquisition of C. neoformans remain poorly defined. In this study, we found that during acute C. neoformans infection, CCR2+ Ly6Chi inflammatory monocytes (IM) rapidly infiltrate the lungs and mediate fungal trafficking to lung-draining lymph nodes. Interestingly, this influx of IM is detrimental to the host, since ablating IM or impairing their recruitment to the lungs improves murine survival and reduces fungal proliferation and dissemination. Using a novel conditional gene deletion strategy, we determined that MHC class II expression by IM did not mediate their deleterious impact on the host. Furthermore, although ablation of IM reduced the number of lymphocytes, innate lymphoid cells, and eosinophils in the lungs, the effects of IM were not dependent on these cells. We ascertained that IM in the lungs upregulated transcripts associated with alternatively activated (M2) macrophages in response to C. neoformans, consistent with the model that IM assume a cellular phenotype that is permissive for fungal growth. We also determined that conditional knockout of the prototypical M2 marker arginase 1 in IM and deletion of the M2-associated transcription factor STAT6 were not sufficient to reverse the harmful effects of IM. Overall, our findings indicate that C. neoformans can subvert the fungicidal potential of IM to enable the progression of infection through a mechanism that is not dependent on lymphocyte priming, eosinophil recruitment, or downstream M2 macrophage polarization pathways. These results give us new insight into the plasticity of IM function during fungal infections and the level of control that C. neoformans can exert on host immune responses. Cryptococcus neoformans is a fungus that is prevalent throughout the environment and can cause a fatal infection of the central nervous system when inhaled into the lungs by patients with impaired immune systems. Our understanding of the immune responses that either help clear C. neoformans from the lungs or permit development of disease remains limited. In this study, we used a mouse model of lethal C. neoformans infection to determine that inflammatory monocytes, immune cells that are often among the first responders to infections, actually facilitate the progression of infection rather than clearance. These findings establish a foundation for future work to target the immune response of inflammatory monocytes as a strategy to improve the outcomes of patients that develop C. neoformans infections.
Collapse
Affiliation(s)
- Lena J. Heung
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail: (LJH); (TMH)
| | - Tobias M. Hohl
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail: (LJH); (TMH)
| |
Collapse
|
36
|
Interleukin-Mediated Pendrin Transcriptional Regulation in Airway and Esophageal Epithelia. Int J Mol Sci 2019; 20:ijms20030731. [PMID: 30744098 PMCID: PMC6386862 DOI: 10.3390/ijms20030731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
Pendrin (SLC26A4), a Cl−/anion exchanger, is expressed at high levels in kidney, thyroid, and inner ear epithelia, where it has an essential role in bicarbonate secretion/chloride reabsorption, iodide accumulation, and endolymph ion balance, respectively. Pendrin is expressed at lower levels in other tissues, such as airways and esophageal epithelia, where it is transcriptionally regulated by the inflammatory cytokines interleukin (IL)-4 and IL-13 through a signal transducer and activator of transcription 6 (STAT6)-mediated pathway. In the airway epithelium, increased pendrin expression during inflammatory diseases leads to imbalances in airway surface liquid thickness and mucin release, while, in the esophageal epithelium, dysregulated pendrin expression is supposed to impact the intracellular pH regulation system. In this review, we discuss some of the recent findings on interleukin-mediated transcriptional regulation of pendrin and how this dysregulation impacts airway and esophagus epithelial homeostasis during inflammatory diseases.
Collapse
|
37
|
Src promotes anti-inflammatory (M2) macrophage generation via the IL-4/STAT6 pathway. Cytokine 2018; 111:209-215. [DOI: 10.1016/j.cyto.2018.08.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/19/2018] [Accepted: 08/28/2018] [Indexed: 01/12/2023]
|
38
|
Veremeyko T, Yung AWY, Anthony DC, Strekalova T, Ponomarev ED. Early Growth Response Gene-2 Is Essential for M1 and M2 Macrophage Activation and Plasticity by Modulation of the Transcription Factor CEBPβ. Front Immunol 2018; 9:2515. [PMID: 30443252 PMCID: PMC6221966 DOI: 10.3389/fimmu.2018.02515] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 10/11/2018] [Indexed: 12/24/2022] Open
Abstract
The process of macrophage polarization is involved in many pathologies such as anti-cancer immunity and autoimmune diseases. Polarized macrophages exhibit various levels of plasticity when M2/M(IL-4) macrophages are reprogrammed into an M1-like phenotype following treatment with IFNγ and/or LPS. At the same time, M1 macrophages are resistant to reprogramming in the presence of M2-like stimuli. The molecular mechanisms responsible for the macrophages polarization, plasticity of M2 macrophages, and lack of plasticity in M1 macrophages remain unknown. Here, we explored the role of Egr2 in the induction and maintenance of macrophage M1 and M2 polarization in the mouse in vitro and in vivo models of inflammation. Egr2 knockdown with siRNA treatment fail to upregulate either M1 or M2 markers upon stimulation, and the overexpression of Egr2 potentiated M1 or M2 marker expression following polarization. Polarisation with M2-like stimuli (IL-4 or IL-13) results in increased Egr2 expression, but macrophages stimulated with M1-like stimuli (IFNγ, LPS, IL-6, or TNF) exhibit a decrease in Egr2 expression. Egr2 was critical for the expression of transcription factors CEBPβ and PPARγ in M2 macrophages, and CEBPβ was highly expressed in M1-polarized macrophages. In siRNA knockdown studies the transcription factor CEBPβ was found to negatively regulate Egr2 expression and is likely to be responsible for the maintenance of the M1-like phenotype and lack plasticity. During thioglycolate-induced peritonitis, adoptively transferred macrophages with Egr2 knockdown failed to become activated as determined by upregulation of MHC class II and CD86. Thus, our study indicates that Egr2 expression is associated with the ability of unstimulated or M2 macrophages to respond to stimulation with inflammatory stimuli, while low levels of Egr2 expression is associated with non-responsiveness of macrophages to their activation.
Collapse
Affiliation(s)
- Tatyana Veremeyko
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Amanda W Y Yung
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Tatyana Strekalova
- Department of Neuroscience, Maastricht University, Maastricht, Netherlands.,Institute of General Pathology and Pathophysiology, Moscow, Russia.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Eugene D Ponomarev
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong.,Kunming Institute of Zoology-Chinese University of Hong Kong Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming, China
| |
Collapse
|
39
|
Smith H, Forman R, Mair I, Else KJ. Interactions of helminths with macrophages: therapeutic potential for inflammatory intestinal disease. Expert Rev Gastroenterol Hepatol 2018; 12:997-1006. [PMID: 30113218 DOI: 10.1080/17474124.2018.1505498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Macrophages represent a highly heterogeneous and plastic cell type found in most tissues of the body; the intestine is home to enormous numbers of these cells. Considerable interest surrounds the 'M2 macrophage,' as it is able to control and regulate inflammation, while promoting tissue repair. Areas covered: As potent inducers of M2 macrophages, intestinal helminths and helminth-derived products are ideal candidates for small molecule drug design to drive M2 macrophage polarization. Several gastrointestinal helminths have been found to cause M2 macrophage-inducing infections. This review covers current knowledge of helminth products and their impact on macrophage polarization, which may in the future lead to new therapeutic strategies. A literature search was performed using the following search terms in PubMed: M2 macrophage, alternative activation, helminth products, helminth ES, helminth therapy, nanoparticle, intestinal macrophages. Other studies were selected by using references from articles identified through our original literature search. Expert commentary: While the immunomodulatory potential of helminth products is well established, we have yet to fully characterize many components of the intestinal helminth product library. Current work aims to identify the protein motifs responsible for modulation of macrophages and other components of the immune system.
Collapse
Affiliation(s)
- Hannah Smith
- a Faculty of Biology, Medicine and Health , University of Manchester , Manchester , UK.,b Manchester Academic Health Sciences Centre , Manchester , UK
| | - Ruth Forman
- a Faculty of Biology, Medicine and Health , University of Manchester , Manchester , UK.,b Manchester Academic Health Sciences Centre , Manchester , UK
| | - Iris Mair
- a Faculty of Biology, Medicine and Health , University of Manchester , Manchester , UK.,b Manchester Academic Health Sciences Centre , Manchester , UK
| | - Kathryn J Else
- a Faculty of Biology, Medicine and Health , University of Manchester , Manchester , UK.,b Manchester Academic Health Sciences Centre , Manchester , UK
| |
Collapse
|
40
|
Choudry M, Tang X, Santorian T, Wasnik S, Xiao J, Xing W, Lau KW, Mohan S, Baylink DJ, Qin X. Deficient arginase II expression without alteration in arginase I expression attenuated experimental autoimmune encephalomyelitis in mice. Immunology 2018; 155:85-98. [PMID: 29574762 PMCID: PMC6099175 DOI: 10.1111/imm.12926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/24/2018] [Accepted: 02/25/2018] [Indexed: 01/02/2023] Open
Abstract
In the past there have been a multitude of studies that ardently support the role of arginase II (Arg II) in vascular and endothelial disorders; however, the regulation and function of Arg II in autoimmune diseases has thus far remained unclear. Here we report that a global Arg II null mutation in mice suppressed experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. During EAE, both Arg I and Arg II were induced in spinal cords, but only Arg II was induced in spleens and splenic dendritic cells (DCs). DC activation by lipopolysaccharide (LPS), CD40L or TLR8 agonist significantly enhanced Arg II expression without affecting Arg I expression. Conversely, DC differentiating cytokines [IL-4 and granulocyte macrophage-colony-stimulating factor (GM-CSF)] yielded opposite effects. In addition, Arg I and Arg II were regulated differentially during Th1 and Th17 cell polarization. Arg II deficiency in mice delayed EAE onset, ameliorated clinical symptoms and reduced myelin loss, accompanied by a remarkable reduction in the EAE-induced spinal cord expression of Th17 cell markers (IL-17 and RORγt). The abundance of Th17 cells and IL-23+ cells in relevant draining lymph nodes was significantly reduced in Arg II knockout mice. In activated DCs, Arg II deficiency significantly suppressed the expression of Th17-differentiating cytokines IL-23 and IL-6. Interestingly, Arg II deficiency did not lead to any compensatory increase in Arg I expression in vivo and in vitro. In conclusion, Arg II was identified as a factor promoting EAE likely via an Arg I-independent mechanism. Arg II may promote EAE by enhancing DC production of Th17-differentiating cytokines. Specific inhibition of Arg II could be a potential therapy for multiple sclerosis.
Collapse
Affiliation(s)
| | - Xiaolei Tang
- Department of MedicineLoma Linda University School of MedicineLoma LindaCAUSA
| | | | - Samiksha Wasnik
- Department of MedicineLoma Linda University School of MedicineLoma LindaCAUSA
| | - Jidong Xiao
- Department of Ultrasound & ImagingThird Xiangya HospitalCentral South UniversityChangshaChina
| | - Weirong Xing
- J. L Pettis VA Medical CenterLoma LindaCAUSA
- Department of MedicineLoma Linda University School of MedicineLoma LindaCAUSA
| | - Kin‐Hing William Lau
- J. L Pettis VA Medical CenterLoma LindaCAUSA
- Department of MedicineLoma Linda University School of MedicineLoma LindaCAUSA
| | - Subburaman Mohan
- J. L Pettis VA Medical CenterLoma LindaCAUSA
- Department of MedicineLoma Linda University School of MedicineLoma LindaCAUSA
| | - David J. Baylink
- Department of MedicineLoma Linda University School of MedicineLoma LindaCAUSA
| | - Xuezhong Qin
- J. L Pettis VA Medical CenterLoma LindaCAUSA
- Department of MedicineLoma Linda University School of MedicineLoma LindaCAUSA
| |
Collapse
|
41
|
Zhang Y, Wu L, Li Z, Zhang W, Luo F, Chu Y, Chen G. Glycocalyx-Mimicking Nanoparticles Improve Anti-PD-L1 Cancer Immunotherapy through Reversion of Tumor-Associated Macrophages. Biomacromolecules 2018; 19:2098-2108. [DOI: 10.1021/acs.biomac.8b00305] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yufei Zhang
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| | - Libin Wu
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| | - Zhen Li
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| | - Weiyi Zhang
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Guosong Chen
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| |
Collapse
|
42
|
Giles DA, Washnock-Schmid JM, Duncker PC, Dahlawi S, Ponath G, Pitt D, Segal BM. Myeloid cell plasticity in the evolution of central nervous system autoimmunity. Ann Neurol 2018; 83:131-141. [PMID: 29283442 PMCID: PMC5876132 DOI: 10.1002/ana.25128] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/22/2017] [Accepted: 12/26/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Myeloid cells, including macrophages and dendritic cells, are a prominent component of central nervous system (CNS) infiltrates during multiple sclerosis (MS) and the animal model experimental autoimmune encephalomyelitis (EAE). Although myeloid cells are generally thought to be proinflammatory, alternatively polarized subsets can serve noninflammatory and/or reparative functions. Here we investigate the heterogeneity and biological properties of myeloid cells during central nervous system autoimmunity. METHODS Myeloid cell phenotypes in chronic active MS lesions were analyzed by immunohistochemistry. In addition, immune cells were isolated from the CNS during exacerbations and remissions of EAE and characterized by flow cytometric, genetic, and functional assays. RESULTS Myeloid cells expressing inducible nitric oxide synthase (iNOS), indicative of a proinflammatory phenotype, were detected in the actively demyelinating rim of chronic active MS lesions, whereas macrophages expressing mannose receptor (CD206), a marker of alternatively polarized human myeloid cells, were enriched in the quiescent lesion core. During EAE, CNS-infiltrating myeloid cells, as well as microglia, shifted from expression of proinflammatory markers to expression of noninflammatory markers immediately prior to clinical remissions. Murine CNS myeloid cells expressing the alternative lineage marker arginase-1 (Arg1) were partially derived from iNOS+ precursors and were deficient in activating encephalitogenic T cells compared with their Arg1- counterparts. INTERPRETATION These observations demonstrate the heterogeneity of CNS myeloid cells, their evolution during the course of autoimmune demyelinating disease, and their plasticity on the single cell level. Future therapeutic strategies for disease modification in individuals with MS may be focused on accelerating the transition of CNS myeloid cells from a proinflammatory to a noninflammatory phenotype. Ann Neurol 2018;83:131-141.
Collapse
Affiliation(s)
- David A. Giles
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Jesse M. Washnock-Schmid
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Patrick C. Duncker
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Somiah Dahlawi
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - Gerald Ponath
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - David Pitt
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - Benjamin M. Segal
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
43
|
Lipid bodies containing oxidatively truncated lipids block antigen cross-presentation by dendritic cells in cancer. Nat Commun 2017; 8:2122. [PMID: 29242535 PMCID: PMC5730553 DOI: 10.1038/s41467-017-02186-9] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 11/08/2017] [Indexed: 01/15/2023] Open
Abstract
Cross-presentation is a critical function of dendritic cells (DCs) required for induction of antitumor immune responses and success of cancer immunotherapy. It is established that tumor-associated DCs are defective in their ability to cross-present antigens. However, the mechanisms driving these defects are still unknown. We find that impaired cross-presentation in DCs is largely associated with defect in trafficking of peptide-MHC class I (pMHC) complexes to the cell surface. DCs in tumor-bearing hosts accumulate lipid bodies (LB) containing electrophilic oxidatively truncated (ox-tr) lipids. These ox-tr-LB, but not LB present in control DCs, covalently bind to chaperone heat shock protein 70. This interaction prevents the translocation of pMHC to cell surface by causing the accumulation of pMHC inside late endosomes/lysosomes. As a result, tumor-associated DCs are no longer able to stimulate adequate CD8 T cells responses. In conclusion, this study demonstrates a mechanism regulating cross-presentation in cancer and suggests potential therapeutic avenues.
Collapse
|
44
|
Barilli A, Gaiani F, Prandi B, Cirlini M, Ingoglia F, Visigalli R, Rotoli BM, de'Angelis N, Sforza S, de'Angelis GL, Dall'Asta V. Gluten peptides drive healthy and celiac monocytes toward an M2-like polarization. J Nutr Biochem 2017; 54:11-17. [PMID: 29216605 DOI: 10.1016/j.jnutbio.2017.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 10/20/2017] [Accepted: 10/26/2017] [Indexed: 01/16/2023]
Abstract
Celiac disease (CD) is an immune-mediated enteropathy triggered by ingested gluten in genetically susceptible individuals and sustained by both adaptive and innate immune responses. Recent studies in murine macrophages demonstrated that the activation of arginase (ARG) metabolic pathway by gluten peptides contributes to the modulation of intestinal permeability in vitro. Here we characterize the effects of gluten on arginine metabolism and cell polarization in human monocytes from both healthy and CD subjects; both a simplified enzymatic digestion of gliadin and a physiological digestion of whole wheat have been tested. Results indicate that gluten digests induce the onset of an M2-like phenotype in activated macrophages; more precisely, both isoforms of arginase, ARG1 and ARG2, are induced likely due to the inhibition of mTOR and the consequent induction of C/EBPβ transcription factor. These effects are independent from the origin of gluten as well as from the digestive protocol employed; moreover, no statistical difference can be evidenced between healthy and CD patients, excluding a diverse predisposition of CD monocytes to gluten-triggered polarization with respect to healthy immune cells. Overall, the present findings sustain a role for arginase pathway in the immune response elicited by human monocytes toward ingested gluten that, hence, deserves particular attention when addressing the pathogenesis of CD.
Collapse
Affiliation(s)
- Amelia Barilli
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Federica Gaiani
- Unit of Gastroenterology and Digestive Endoscopy of Parma, University Hospital of Parma, via Gramsci 14, 43126 Parma, Italy
| | - Barbara Prandi
- Department of Food Science, University of Parma, Parco Area delle Scienze 17A, 43124 Parma, Italy
| | - Martina Cirlini
- Department of Food Science, University of Parma, Parco Area delle Scienze 17A, 43124 Parma, Italy
| | - Filippo Ingoglia
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Rossana Visigalli
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Bianca Maria Rotoli
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy
| | - Nicola de'Angelis
- Department of HPB Surgery and Liver Transplantation, Henri-Mondor Hospital, Université Paris Est-UPEC, Créteil, France
| | - Stefano Sforza
- Department of Food Science, University of Parma, Parco Area delle Scienze 17A, 43124 Parma, Italy
| | - Gian Luigi de'Angelis
- Unit of Gastroenterology and Digestive Endoscopy of Parma, University Hospital of Parma, via Gramsci 14, 43126 Parma, Italy
| | - Valeria Dall'Asta
- Unit of General Pathology, Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
45
|
Murray PJ. Nonresolving macrophage-mediated inflammation in malignancy. FEBS J 2017; 285:641-653. [PMID: 28857430 DOI: 10.1111/febs.14210] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/07/2017] [Accepted: 08/25/2017] [Indexed: 12/14/2022]
Abstract
Tumors are populated with different cells of the immune system, each of which has the potential for pro- or antitumor functions. Macrophages are the numerically dominant type of myeloid cell in cancer and are suspected of having predominantly protumor functions. Key questions in cancer research concern the relationships between macrophages and anatomically different kinds of cancers, what specific properties of macrophages are involved in protumor functions and whether either macrophage numbers or functions can be modulated to enhance existing cancer therapies, for example, by reducing the immunosuppressive milieu such that anti-tumor T cells can provoke antitumor immunity. Accordingly, several antimacrophage preclinical modalities have been attempted and revealed substantial clinical barriers to their use. Therefore, understanding and targeting the specific pathways associated with protumor functions of macrophages, rather than macrophages themselves is a promising approach for both basic research and therapeutic development.
Collapse
Affiliation(s)
- Peter J Murray
- Immunoregulation Group, Max-Planck-Institut für Biochemie, Martinsried, Germany
| |
Collapse
|
46
|
The transcription factor MafB promotes anti-inflammatory M2 polarization and cholesterol efflux in macrophages. Sci Rep 2017; 7:7591. [PMID: 28790455 PMCID: PMC5548719 DOI: 10.1038/s41598-017-07381-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 06/28/2017] [Indexed: 12/21/2022] Open
Abstract
Macrophages play pivotal roles in the progression and regression of atherosclerosis. Accumulating evidence suggests that macrophage polarization into an anti-inflammatory M2 state is a key characteristic of atherosclerotic plaques undergoing regression. However, the molecular mechanisms underlying this potential association of the M2 polarization with atherosclerosis regression remain poorly understood. Further, human genetic factors that facilitate these anti-atherogenic processes remain largely unknown. We report that the transcription factor MafB plays pivotal roles in promoting macrophage M2 polarization. Further, MafB promotes cholesterol efflux from macrophage foam cells by directly up-regulating its key cellular mediators. Notably, MafB expression is significantly up-regulated in response to various metabolic and immunological stimuli that promote macrophage M2 polarization or cholesterol efflux, and thereby MafB mediates their beneficial effects, in both liver x receptor (LXR)-dependent and independent manners. In contrast, MafB is strongly down-regulated upon elevated pro-inflammatory signaling or by pro-inflammatory and pro-atherogenic microRNAs, miR-155 and miR-33. Using an integrative systems biology approach, we also revealed that M2 polarization and cholesterol efflux do not necessarily represent inter-dependent events, but MafB is broadly involved in both the processes. These findings highlight physiological protective roles that MafB may play against atherosclerosis progression.
Collapse
|
47
|
Chen Y, Zhang X. Pivotal regulators of tissue homeostasis and cancer: macrophages. Exp Hematol Oncol 2017; 6:23. [PMID: 28804688 PMCID: PMC5549331 DOI: 10.1186/s40164-017-0083-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022] Open
Abstract
Macrophages are an essential component of innate immunity and play a vital role in inflammation and host defense. Based on immunological responses, the macrophages are classified into "activated" macrophage (M1 macrophages) participating in the responses of type I helper T (Th1) cells to pathogens and "alternatively activated" macrophages (M2 macrophages) in response to interleukin (IL)-4 and IL-13. In this review, we discuss the origin, classification and function of macrophages. We also discuss the mechanisms underlying polarization of different macrophage subtypes, including transcriptional, epigenetic and post-transcriptional regulation.
Collapse
Affiliation(s)
- Yulei Chen
- College of Life Sciences, Zhejiang University, Hangzhou, 310058 People's Republic of China
| | - Xiaobo Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, 310058 People's Republic of China
| |
Collapse
|
48
|
Abstract
SIGNIFICANCE Leukocytes and especially macrophages are a major cellular constituent of the tumor mass. The tumor microenvironment not only determines their activity but in turn these cells also contribute to tumor initiation and progression. Recent Advances: Proinflammatory stimulated macrophages upregulate inducible nitric oxide synthase (NOS2) and produce high steady-state NO concentrations. NO provokes tumor cell death by initiating apoptosis and/or necrosis. Mechanisms may comprise p53 accumulation, immunestimulatory activities, and an increased efficacy of chemo- and/or radiotherapy. However, the potential cytotoxic activity of macrophages often is compromised in the tumor microenvironment and instead a protumor activity of macrophages dominates. Contributing factors are signals generated by viable and dying tumor cells, attraction and activation of myeloid-derived suppressor cells, and hypoxia. Limited oxygen availability not only attenuates NOS2 activity but also causes accumulation of hypoxia-inducible factors 1 and 2 (HIF-1/HIF-2). Activation of the HIF system is tightly linked to NO formation and affects the expression of macrophage phenotype markers that in turn add to tumor progression. CRITICAL ISSUES To make use of the cytotoxic arsenal of activated macrophages directed against tumor cells, it will be critical to understand how, when, and where these innate immune responses are blocked and whether it will be possible to reinstall their full capacity to kill tumor cells. FUTURE DIRECTIONS Low-dose irradiation or proinflammatory activation of macrophages in the tumor microenvironment may open options to boost NOS2 expression and activity and to initiate immunestimulatory features of NO that may help to restrict tumor growth. Antioxid. Redox Signal. 26, 1023-1043.
Collapse
Affiliation(s)
- Bernhard Brüne
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt , Frankfurt, Germany
| | - Nadine Courtial
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt , Frankfurt, Germany
| | - Nathalie Dehne
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt , Frankfurt, Germany
| | - Shahzad N Syed
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt , Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt , Frankfurt, Germany
| |
Collapse
|
49
|
Wilson NM, Gurney ME, Dietrich WD, Atkins CM. Therapeutic benefits of phosphodiesterase 4B inhibition after traumatic brain injury. PLoS One 2017; 12:e0178013. [PMID: 28542295 PMCID: PMC5438188 DOI: 10.1371/journal.pone.0178013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/05/2017] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) initiates a deleterious inflammatory response that exacerbates pathology and worsens outcome. This inflammatory response is partially mediated by a reduction in cAMP and a concomitant upregulation of cAMP-hydrolyzing phosphodiesterases (PDEs) acutely after TBI. The PDE4B subfamily, specifically PDE4B2, has been found to regulate cAMP in inflammatory cells, such as neutrophils, macrophages and microglia. To determine if PDE4B regulates inflammation and subsequent pathology after TBI, adult male Sprague Dawley rats received sham surgery or moderate parasagittal fluid-percussion brain injury (2 ± 0.2 atm) and were then treated with a PDE4B - selective inhibitor, A33, or vehicle for up to 3 days post-surgery. Treatment with A33 reduced markers of microglial activation and neutrophil infiltration at 3 and 24 hrs after TBI, respectively. A33 treatment also reduced cortical contusion volume at 3 days post-injury. To determine whether this treatment paradigm attenuated TBI-induced behavioral deficits, animals were evaluated over a period of 6 weeks after surgery for forelimb placement asymmetry, contextual fear conditioning, water maze performance and spatial working memory. A33 treatment significantly improved contextual fear conditioning and water maze retention at 24 hrs post-training. However, this treatment did not rescue sensorimotor or working memory deficits. At 2 months after surgery, atrophy and neuronal loss were measured. A33 treatment significantly reduced neuronal loss in the pericontusional cortex and hippocampal CA3 region. This treatment paradigm also reduced cortical, but not hippocampal, atrophy. Overall, these results suggest that acute PDE4B inhibition may be a viable treatment to reduce inflammation, pathology and memory deficits after TBI.
Collapse
Affiliation(s)
- Nicole M. Wilson
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Mark E. Gurney
- Tetra Discovery Partners, Grand Rapids, Michigan, United States of America
| | - W. Dalton Dietrich
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Coleen M. Atkins
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| |
Collapse
|
50
|
Grohmann U, Mondanelli G, Belladonna ML, Orabona C, Pallotta MT, Iacono A, Puccetti P, Volpi C. Amino-acid sensing and degrading pathways in immune regulation. Cytokine Growth Factor Rev 2017; 35:37-45. [PMID: 28545736 DOI: 10.1016/j.cytogfr.2017.05.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 02/07/2023]
Abstract
Indoleamine 2,3-dioxygenases (IDOs) - belonging in the heme dioxygenase family and degrading tryptophan - are responsible for the de novo synthesis of nicotinamide adenine dinucleotide (NAD+). As such, they are expressed by a variety of invertebrate and vertebrate species. In mammals, IDO1 has remarkably evolved to expand its functions, so to become a prominent homeostatic regulator, capable of modulating infection and immunity in multiple ways, including local tryptophan deprivation, production of biologically active tryptophan catabolites, and non-enzymatic cell-signaling activity. Much like IDO1, arginase 1 (Arg1) is an immunoregulatory enzyme that catalyzes the degradation of arginine. Here, we discuss the possible role of amino-acid degradation as related to the evolution of the immune systems and how the functions of those enzymes are linked by an entwined pathway selected by phylogenesis to meet the newly arising needs imposed by an evolving environment.
Collapse
Affiliation(s)
- Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Maria L Belladonna
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Ciriana Orabona
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Maria T Pallotta
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Alberta Iacono
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| | - Claudia Volpi
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
| |
Collapse
|