1
|
Karimova AF, Khalitova AR, Suezov R, Markov N, Mukhamedshina Y, Rizvanov AA, Huber M, Simon HU, Brichkina A. Immunometabolism of tumor-associated macrophages: A therapeutic perspective. Eur J Cancer 2025; 220:115332. [PMID: 40048925 DOI: 10.1016/j.ejca.2025.115332] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 04/26/2025]
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in the tumor microenvironment (TME), actively contributing to the formation of an immunosuppressive niche that fosters tumor progression. Consequently, there has been a growing interest in targeting TAMs as a promising avenue for cancer therapy. Recent advances in the field of immunometabolism have shed light on the influence of metabolic adaptations on macrophage physiology in the context of cancer. Here, we discuss the key metabolic pathways that shape the phenotypic diversity of macrophages. We place special emphasis on how metabolic reprogramming impacts the activation status of TAMs and their functions within the TME. Additionally, we explore alterations in TAM metabolism and their effects on phagocytosis, production of cytokines/chemokines and interaction with cytotoxic T and NK immune cells. Moreover, we examine the application of nanomedical approaches to target TAMs and assess the clinical significance of modulating the metabolism of TAMs as a strategy to develop new anti-cancer therapies. Taken together, in this comprehensive review article focusing on TAMs, we provide invaluable insights for the development of effective immunotherapeutic strategies and the enhancement of clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Adelya F Karimova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Adelya R Khalitova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Roman Suezov
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany
| | - Nikita Markov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Yana Mukhamedshina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Magdalena Huber
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany
| | - Hans-Uwe Simon
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Institute of Pharmacology, University of Bern, Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Anna Brichkina
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany.
| |
Collapse
|
2
|
Desharnais L, Sorin M, Rezanejad M, Liu B, Karimi E, Atallah A, Swaby AM, Yu MW, Doré S, Hartner S, Fiset B, Wei Y, Kadang B, Rayes R, Joubert P, Camilleri-Broët S, Fiset PO, Quail DF, Spicer JD, Walsh LA. Spatially mapping the tumour immune microenvironments of non-small cell lung cancer. Nat Commun 2025; 16:1345. [PMID: 39905080 PMCID: PMC11794701 DOI: 10.1038/s41467-025-56546-x] [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: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths. An enhanced understanding of the immune microenvironments within these tumours may foster more precise and efficient treatment, particularly for immune-targeted therapies. The spatial architectural differences between lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are relatively unexplored. Here, we applied imaging mass cytometry to a balanced cohort of LUAD and LUSC patients, matched for clinical factors such as age, sex, and smoking history, to analyze 204 histopathology images of tumours from 102 individuals with non-small cell lung cancer (NSCLC). By analyzing interactions and broader cellular networks, we interrogate the tumour microenvironment to understand how immune cells are spatially organized in clinically matched adenocarcinoma and squamous cell carcinoma subsets. This spatial analysis revealed distinct patterns of immune cell aggregation, particularly among macrophage populations, that correlated with patient prognosis differentially in adenocarcinoma and squamous cell carcinoma, suggesting potential new strategies for therapeutic intervention. Our findings underscore the importance of analyzing NSCLC histological subtypes separately when investigating the spatial immune landscape, as microenvironmental characteristics and cellular interactions differed by subtype. Recognizing these distinctions is essential for designing precision therapies tailored to each subtype's unique immune architecture, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Lysanne Desharnais
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Mark Sorin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Morteza Rezanejad
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Bridget Liu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Aline Atallah
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Anikka M Swaby
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Miranda W Yu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Samuel Doré
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Saskia Hartner
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Yuhong Wei
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Baharak Kadang
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Roni Rayes
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Philippe Joubert
- Département de biologie moléculaire, biochimie médicale et de pathologie, Laval University, Québec City, QC, Canada
| | | | | | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Jonathan D Spicer
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Surgery, McGill University Health Center, Montreal, QC, Canada.
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
3
|
Vitorakis N, Gargalionis AN, Papavassiliou KA, Adamopoulos C, Papavassiliou AG. Precision Targeting Strategies in Pancreatic Cancer: The Role of Tumor Microenvironment. Cancers (Basel) 2024; 16:2876. [PMID: 39199647 PMCID: PMC11352254 DOI: 10.3390/cancers16162876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 09/01/2024] Open
Abstract
Pancreatic cancer demonstrates an ever-increasing incidence over the last years and represents one of the top causes of cancer-associated mortality. Cells of the tumor microenvironment (TME) interact with cancer cells in pancreatic ductal adenocarcinoma (PDAC) tumors to preserve cancer cells' metabolism, inhibit drug delivery, enhance immune suppression mechanisms and finally develop resistance to chemotherapy and immunotherapy. New strategies target TME genetic alterations and specific pathways in cell populations of the TME. Complex molecular interactions develop between PDAC cells and TME cell populations including cancer-associated fibroblasts, myeloid-derived suppressor cells, pancreatic stellate cells, tumor-associated macrophages, tumor-associated neutrophils, and regulatory T cells. In the present review, we aim to fully explore the molecular landscape of the pancreatic cancer TME cell populations and discuss current TME targeting strategies to provide thoughts for further research and preclinical testing.
Collapse
Affiliation(s)
- Nikolaos Vitorakis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios N Gargalionis
- Department of Clinical Biochemistry, 'Attikon' University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Kostas A Papavassiliou
- First University Department of Respiratory Medicine, 'Sotiria' Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
4
|
Bonilla ME, Radyk MD, Perricone MD, Elhossiny AM, Harold AC, Medina-Cabrera PI, Kadiyala P, Shi J, Frankel TL, Carpenter ES, Green MD, Mitrea C, Lyssiotis CA, Pasca di Magliano M. Metabolic landscape of the healthy pancreas and pancreatic tumor microenvironment. JCI Insight 2024; 9:e180114. [PMID: 39315547 PMCID: PMC11457849 DOI: 10.1172/jci.insight.180114] [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: 09/25/2024] Open
Abstract
Pancreatic cancer, one of the deadliest human malignancies, is characterized by a fibro-inflammatory tumor microenvironment and wide array of metabolic alterations. To comprehensively map metabolism in a cell type-specific manner, we harnessed a unique single-cell RNA-sequencing dataset of normal human pancreata. This was compared with human pancreatic cancer samples using a computational pipeline optimized for this study. In the cancer cells we observed enhanced biosynthetic programs. We identified downregulation of mitochondrial programs in several immune populations, relative to their normal counterparts in healthy pancreas. Although granulocytes, B cells, and CD8+ T cells all downregulated oxidative phosphorylation, the mechanisms by which this occurred were cell type specific. In fact, the expression pattern of the electron transport chain complexes was sufficient to identify immune cell types without the use of lineage markers. We also observed changes in tumor-associated macrophage (TAM) lipid metabolism, with increased expression of enzymes mediating unsaturated fatty acid synthesis and upregulation in cholesterol export. Concurrently, cancer cells exhibited upregulation of lipid/cholesterol receptor import. We thus identified a potential crosstalk whereby TAMs provide cholesterol to cancer cells. We suggest that this may be a new mechanism boosting cancer cell growth and a therapeutic target in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jiaqi Shi
- Rogel Cancer Center
- Department of Pathology
| | | | - Eileen S. Carpenter
- Rogel Cancer Center
- Department of Internal Medicine, Division of Gastroenterology and Hepatology
| | - Michael D. Green
- Program in Cancer Biology
- Rogel Cancer Center
- Department of Radiation Oncology; and
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | | | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology
- Rogel Cancer Center
- Department of Internal Medicine, Division of Gastroenterology and Hepatology
| | - Marina Pasca di Magliano
- Rogel Cancer Center
- Department of Surgery
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Zheng W, Yu W, Hua R, He J, Wu N, Tian S, Huang W, Qin L. Systematic analysis of TREM2 and its carcinogenesis in pancreatic cancer. Transl Cancer Res 2024; 13:3200-3216. [PMID: 39145088 PMCID: PMC11319948 DOI: 10.21037/tcr-24-201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/09/2024] [Indexed: 08/16/2024]
Abstract
Background Triggering receptor expressed on myeloid cells 2 (TREM2), a transmembrane immunoglobulin-superfamily receptor, is expressed primarily on cells such as macrophages and dendritic cells. TREM2 has been shown to be associated with diseases such as neurodegeneration, fatty liver, obesity, and atherosclerosis. Currently, it has become one of the hotspots in oncology research. However, the role of TREM2 in pan-cancer, especially pancreatic cancer, remains unclear. Methods We used the Tumor-immune System Interactions Database (TISIDB) to explore TREM2 expression differences, Tumor Immune Single-cell Hub 2 (TISCH2) to explore TREM2 expression distribution, Tumor IMmune Estimation Resource 2.0 (TIMER 2.0) to explore immune infiltration, cBio Cancer Genomics Portal (cBioPortal) to explore genetic variation, Genomics of Drug Sensitivity in Cancer (GDSC) to explore drug resistance, and Kaplan-Meier plotter database to explore the relationship between TREM2 and prognosis in pancreatic cancer. In addition, we used The Cancer Genome Atlas-pancreatic adenocarcinoma (TCGA-PAAD) and normal pancreas samples from the Genotype-Tissue Expression (GTEx) databases to explore the relationship between TREM2 and lymph node metastasis. We verified the protein level of TREM2 in pancreatic cancer by Human Protein Atlas (HPA) and western blotting and detected the colocalization of TREM2 with malignant cell markers by multiplex immunohistochemistry (mIHC). Finally, we identified the tumor-promoting role of TREM2 in pancreatic cancer via in vitro experiments, such as cell cycle assays, colony formation assays, and transwell migration and invasion assays. Results Our results showed that TREM2 was differentially expressed in various tumors according to different molecular and immune subtypes of pan-cancer. It was found that TREM2 was mainly expressed in monocytes/macrophages. In addition, our study showed that TREM2 expression was closely associated with macrophages in the tumor microenvironment (TME) of pan-cancer. TREM2 was shown to be related to anti-inflammatory and immunosuppressive effects in most cancers. Furthermore, we found that amplification was the main somatic mutation of TREM2 in pan-cancer. Further correlational analysis revealed a significant negative association of TREM2 expression with sensitivity to AZD8186, which is a selective inhibitor of PI3K, but not gemcitabine and paclitaxel. Finally, through the knockdown and overexpression of TREM2, our findings verified that TREM2 on cancer cells promoted the progression of PAAD. Conclusions In conclusion, our comprehensive analysis identified that TREM2 expression level was correlated with the TME and the immunosuppressive effects. In particular, our study indicated that TREM2 was involved in the progression of pancreatic cancer.
Collapse
Affiliation(s)
- Wanting Zheng
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Wangjianfei Yu
- Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Ruheng Hua
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Jun He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Nuwa Wu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Siyun Tian
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Wentao Huang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| |
Collapse
|
6
|
Tharp KM, Kersten K, Maller O, Timblin GA, Stashko C, Canale FP, Menjivar RE, Hayward MK, Berestjuk I, Ten Hoeve J, Samad B, Ironside AJ, di Magliano MP, Muir A, Geiger R, Combes AJ, Weaver VM. Tumor-associated macrophages restrict CD8 + T cell function through collagen deposition and metabolic reprogramming of the breast cancer microenvironment. NATURE CANCER 2024; 5:1045-1062. [PMID: 38831058 DOI: 10.1038/s43018-024-00775-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Tumor progression is accompanied by fibrosis, a condition of excessive extracellular matrix accumulation, which is associated with diminished antitumor immune infiltration. Here we demonstrate that tumor-associated macrophages (TAMs) respond to the stiffened fibrotic tumor microenvironment (TME) by initiating a collagen biosynthesis program directed by transforming growth factor-β. A collateral effect of this programming is an untenable metabolic milieu for productive CD8+ T cell antitumor responses, as collagen-synthesizing macrophages consume environmental arginine, synthesize proline and secrete ornithine that compromises CD8+ T cell function in female breast cancer. Thus, a stiff and fibrotic TME may impede antitumor immunity not only by direct physical exclusion of CD8+ T cells but also through secondary effects of a mechano-metabolic programming of TAMs, which creates an inhospitable metabolic milieu for CD8+ T cells to respond to anticancer immunotherapies.
Collapse
Affiliation(s)
- Kevin M Tharp
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Kelly Kersten
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
| | - Ori Maller
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Greg A Timblin
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Connor Stashko
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Fernando P Canale
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Rosa E Menjivar
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Mary-Kate Hayward
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ilona Berestjuk
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Johanna Ten Hoeve
- UCLA Metabolomics Center, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bushra Samad
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | | | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, Cancer Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Roger Geiger
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Alexis J Combes
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences and Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and The Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Raynor JL, Chi H. Nutrients: Signal 4 in T cell immunity. J Exp Med 2024; 221:e20221839. [PMID: 38411744 PMCID: PMC10899091 DOI: 10.1084/jem.20221839] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/28/2024] Open
Abstract
T cells are integral in mediating adaptive immunity to infection, autoimmunity, and cancer. Upon immune challenge, T cells exit from a quiescent state, followed by clonal expansion and effector differentiation. These processes are shaped by three established immune signals, namely antigen stimulation (Signal 1), costimulation (Signal 2), and cytokines (Signal 3). Emerging findings reveal that nutrients, including glucose, amino acids, and lipids, are crucial regulators of T cell responses and interplay with Signals 1-3, highlighting nutrients as Signal 4 to license T cell immunity. Here, we first summarize the functional importance of Signal 4 and the underlying mechanisms of nutrient transport, sensing, and signaling in orchestrating T cell activation and quiescence exit. We also discuss the roles of nutrients in programming T cell differentiation and functional fitness and how nutrients can be targeted to improve disease therapy. Understanding how T cells respond to Signal 4 nutrients in microenvironments will provide insights into context-dependent functions of adaptive immunity and therapeutic interventions.
Collapse
Affiliation(s)
- Jana L Raynor
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
8
|
Meli AP, Russell GA, Swaminathan S, Weichselbaum L, MacMahon CA, Pernet E, Karo-Atar D, Rogers D, Rochette A, Fontes G, Mandl JN, Divangahi M, Klein OD, Gregorieff A, Stäger S, King IL. Bcl-6 expression by CD4 + T cells determines concomitant immunity and host resistance across distinct parasitic infections. Mucosal Immunol 2023; 16:801-816. [PMID: 37659724 DOI: 10.1016/j.mucimm.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/03/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
Cluster of differentiation (CD4+) T cells consist of multiple subtypes, defined by expression of lineage-specific transcription factors, that contribute to the control of infectious diseases by providing help to immune and nonimmune target cells. In the current study, we examined the role of B cell lymphoma (Bcl)-6, a transcriptional repressor and master regulator of T follicular helper cell differentiation, in T cell-mediated host defense against intestinal and systemic parasitic infections. We demonstrate that while Bcl-6 expression by CD4+ T cells is critical for antibody-mediated protective immunity against secondary infection with the nematode Heligmosoides polygyrus bakeri, it paradoxically compromises worm expulsion during primary infection by limiting the generation of interleukin-10 (IL-10)-producing Gata3+ T helper 2 cells. Enhanced worm expulsion in the absence of Bcl-6 expressing T cells was associated with amplified intestinal goblet cell differentiation and increased generation of alternatively activated macrophages, effects that were reversed by neutralization of IL-10 signals. An increase in IL-10 production by Bcl-6-deficient CD4+ T cells was also evident in the context of systemic Leishmania donovani infection, but in contrast to Heligmosoides polygyrus bakeri infection, compromised T helper 1-mediated liver macrophage activation and increased susceptibility to this distinct parasitic challenge. Collectively, our studies suggest that host defense pathways that protect against parasite superinfection and lethal systemic protozoal infections can be engaged at the cost of compromised primary resistance to well-tolerated helminths.
Collapse
Affiliation(s)
- Alexandre P Meli
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada
| | - Gabriel A Russell
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada
| | | | - Laura Weichselbaum
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Clara A MacMahon
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada
| | - Erwan Pernet
- Research Institute of the McGill University Health Centre, Meakins-Christie Laboratories, Department of Medicine, Montreal, Quebec, Canada
| | - Danielle Karo-Atar
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada
| | - Dakota Rogers
- Department of Physiology and McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Annie Rochette
- Department of Pathology and Cancer Research Program, McGill University, Montreal, Quebec, Canada
| | - Ghislaine Fontes
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada
| | - Judith N Mandl
- McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada; Department of Physiology and McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Maziar Divangahi
- Research Institute of the McGill University Health Centre, Meakins-Christie Laboratories, Department of Medicine, Montreal, Quebec, Canada
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alex Gregorieff
- Department of Pathology and Cancer Research Program, McGill University, Montreal, Quebec, Canada; McGill Regenerative Medicine Network, Montreal, Quebec, Canada
| | | | - Irah L King
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada; McGill Regenerative Medicine Network, Montreal, Quebec, Canada; McGill Centre for Microbiome Research, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Margul D, Yu C, AlHilli MM. Tumor Immune Microenvironment in Gynecologic Cancers. Cancers (Basel) 2023; 15:3849. [PMID: 37568665 PMCID: PMC10417375 DOI: 10.3390/cancers15153849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Gynecologic cancers have varying response rates to immunotherapy due to the heterogeneity of each cancer's molecular biology and features of the tumor immune microenvironment (TIME). This article reviews key features of the TIME and its role in the pathophysiology and treatment of ovarian, endometrial, cervical, vulvar, and vaginal cancer. Knowledge of the role of the TIME in gynecologic cancers has been rapidly developing with a large body of preclinical studies demonstrating an intricate yet dichotomous role that the immune system plays in either supporting the growth of cancer or opposing it and facilitating effective treatment. Many targets and therapeutics have been identified including cytokines, antibodies, small molecules, vaccines, adoptive cell therapy, and bacterial-based therapies but most efforts in gynecologic cancers to utilize them have not been effective. However, with the development of immune checkpoint inhibitors, we have started to see the rapid and successful employment of therapeutics in cervical and endometrial cancer. There remain many challenges in utilizing the TIME, particularly in ovarian cancer, and further studies are needed to identify and validate efficacious therapeutics.
Collapse
Affiliation(s)
| | | | - Mariam M. AlHilli
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cleveland Clinic, Cleveland, OH 44195, USA; (D.M.); (C.Y.)
| |
Collapse
|
10
|
Yang L, Chu Z, Liu M, Zou Q, Li J, Liu Q, Wang Y, Wang T, Xiang J, Wang B. Amino acid metabolism in immune cells: essential regulators of the effector functions, and promising opportunities to enhance cancer immunotherapy. J Hematol Oncol 2023; 16:59. [PMID: 37277776 DOI: 10.1186/s13045-023-01453-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/13/2023] [Indexed: 06/07/2023] Open
Abstract
Amino acids are basic nutrients for immune cells during organ development, tissue homeostasis, and the immune response. Regarding metabolic reprogramming in the tumor microenvironment, dysregulation of amino acid consumption in immune cells is an important underlying mechanism leading to impaired anti-tumor immunity. Emerging studies have revealed that altered amino acid metabolism is tightly linked to tumor outgrowth, metastasis, and therapeutic resistance through governing the fate of various immune cells. During these processes, the concentration of free amino acids, their membrane bound transporters, key metabolic enzymes, and sensors such as mTOR and GCN2 play critical roles in controlling immune cell differentiation and function. As such, anti-cancer immune responses could be enhanced by supplement of specific essential amino acids, or targeting the metabolic enzymes or their sensors, thereby developing novel adjuvant immune therapeutic modalities. To further dissect metabolic regulation of anti-tumor immunity, this review summarizes the regulatory mechanisms governing reprogramming of amino acid metabolism and their effects on the phenotypes and functions of tumor-infiltrating immune cells to propose novel approaches that could be exploited to rewire amino acid metabolism and enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Luming Yang
- Chongqing University Medical School, Chongqing, 400044, People's Republic of China
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Zhaole Chu
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Meng Liu
- Chongqing University Medical School, Chongqing, 400044, People's Republic of China
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Qiang Zou
- Chongqing University Medical School, Chongqing, 400044, People's Republic of China
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Jinyang Li
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Qin Liu
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China
| | - Yazhou Wang
- Chongqing University Medical School, Chongqing, 400044, People's Republic of China.
| | - Tao Wang
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China.
| | - Junyu Xiang
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China.
| | - Bin Wang
- Department of Gastroenterology and Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, People's Republic of China.
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China.
- Jinfeng Laboratory, Chongqing, 401329, People's Republic of China.
| |
Collapse
|
11
|
Han J, Dong L, Wu M, Ma F. Dynamic polarization of tumor-associated macrophages and their interaction with intratumoral T cells in an inflamed tumor microenvironment: from mechanistic insights to therapeutic opportunities. Front Immunol 2023; 14:1160340. [PMID: 37251409 PMCID: PMC10219223 DOI: 10.3389/fimmu.2023.1160340] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/20/2023] [Indexed: 05/31/2023] Open
Abstract
Immunotherapy has brought a paradigm shift in the treatment of tumors in recent decades. However, a significant proportion of patients remain unresponsive, largely due to the immunosuppressive tumor microenvironment (TME). Tumor-associated macrophages (TAMs) play crucial roles in shaping the TME by exhibiting dual identities as both mediators and responders of inflammation. TAMs closely interact with intratumoral T cells, regulating their infiltration, activation, expansion, effector function, and exhaustion through multiple secretory and surface factors. Nevertheless, the heterogeneous and plastic nature of TAMs renders the targeting of any of these factors alone inadequate and poses significant challenges for mechanistic studies and clinical translation of corresponding therapies. In this review, we present a comprehensive summary of the mechanisms by which TAMs dynamically polarize to influence intratumoral T cells, with a focus on their interaction with other TME cells and metabolic competition. For each mechanism, we also discuss relevant therapeutic opportunities, including non-specific and targeted approaches in combination with checkpoint inhibitors and cellular therapies. Our ultimate goal is to develop macrophage-centered therapies that can fine-tune tumor inflammation and empower immunotherapy.
Collapse
Affiliation(s)
- Jiashu Han
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, China
| | - Luochu Dong
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Fei Ma
- Center for National Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Halbrook CJ, Lyssiotis CA, Pasca di Magliano M, Maitra A. Pancreatic cancer: Advances and challenges. Cell 2023; 186:1729-1754. [PMID: 37059070 PMCID: PMC10182830 DOI: 10.1016/j.cell.2023.02.014] [Citation(s) in RCA: 515] [Impact Index Per Article: 257.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/17/2023] [Accepted: 02/08/2023] [Indexed: 04/16/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest cancers. Significant efforts have largely defined major genetic factors driving PDAC pathogenesis and progression. Pancreatic tumors are characterized by a complex microenvironment that orchestrates metabolic alterations and supports a milieu of interactions among various cell types within this niche. In this review, we highlight the foundational studies that have driven our understanding of these processes. We further discuss the recent technological advances that continue to expand our understanding of PDAC complexity. We posit that the clinical translation of these research endeavors will enhance the currently dismal survival rate of this recalcitrant disease.
Collapse
Affiliation(s)
- Christopher J Halbrook
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, CA 92868, USA.
| | - Costas A Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Marina Pasca di Magliano
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Anirban Maitra
- Department of Translational Molecular Pathology, Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Angka L, Tanese de Souza C, Baxter KE, Khan ST, Market M, Martel AB, Tai LH, Kennedy MA, Bell JC, Auer RC. Perioperative arginine prevents metastases by accelerating natural killer cell recovery after surgery. Mol Ther 2022; 30:3270-3283. [PMID: 35619558 PMCID: PMC9552810 DOI: 10.1016/j.ymthe.2022.05.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 05/21/2022] [Accepted: 05/21/2022] [Indexed: 10/18/2022] Open
Abstract
Profound natural killer (NK) cell suppression after cancer surgery is a main driver of metastases and recurrence, for which there is no clinically approved intervention available. Surgical stress is known to cause systemic postoperative changes that negatively modulate NK cell function including the expansion of surgery-induced myeloid-derived suppressor cells (Sx-MDSCs) and a marked reduction in arginine bioavailability. In this study, we determine that Sx-MDSCs regulate systemic arginine levels in the postoperative period and that restoring arginine imbalance after surgery by dietary intake alone was sufficient to significantly reduce surgery-induced metastases in our preclinical murine models. Importantly, the effects of perioperative arginine were dependent upon NK cells. Although perioperative arginine did not prevent immediate NK cell immunoparalysis after surgery, it did accelerate their return to preoperative cytotoxicity, interferon gamma secretion, and activating receptor expression. Finally, in a cohort of patients with colorectal cancer, postoperative arginine levels were shown to correlate with their Sx-MDSC levels. Therefore, this study lends further support for the use of perioperative arginine supplementation by improving NK cell recovery after surgery.
Collapse
Affiliation(s)
- Leonard Angka
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | | | - Katherine E Baxter
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Sarwat T Khan
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Marisa Market
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Andre B Martel
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada; Division of General Surgery, Department of Surgery, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Lee-Hwa Tai
- Department of Immunology & Cell Biology, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Michael A Kennedy
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - John C Bell
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada; Division of General Surgery, Department of Surgery, University of Ottawa, Ottawa, ON K1H 8L1, Canada; Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada.
| |
Collapse
|
14
|
Dichtl S, Sanin DE, Koss CK, Willenborg S, Petzold A, Tanzer MC, Dahl A, Kabat AM, Lindenthal L, Zeitler L, Satzinger S, Strasser A, Mann M, Roers A, Eming SA, El Kasmi KC, Pearce EJ, Murray PJ. Gene-selective transcription promotes the inhibition of tissue reparative macrophages by TNF. Life Sci Alliance 2022; 5:5/4/e202101315. [PMID: 35027468 PMCID: PMC8761491 DOI: 10.26508/lsa.202101315] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/24/2022] Open
Abstract
Pro-inflammatory TNF is a highly gene-selective inhibitor of the gene expression program of tissue repair and wound healing macrophages. Anti-TNF therapies are a core anti-inflammatory approach for chronic diseases such as rheumatoid arthritis and Crohn’s Disease. Previously, we and others found that TNF blocks the emergence and function of alternative-activated or M2 macrophages involved in wound healing and tissue-reparative functions. Conceivably, anti-TNF drugs could mediate their protective effects in part by an altered balance of macrophage activity. To understand the mechanistic basis of how TNF regulates tissue-reparative macrophages, we used RNAseq, scRNAseq, ATACseq, time-resolved phospho-proteomics, gene-specific approaches, metabolic analysis, and signaling pathway deconvolution. We found that TNF controls tissue-reparative macrophage gene expression in a highly gene-specific way, dependent on JNK signaling via the type 1 TNF receptor on specific populations of alternative-activated macrophages. We further determined that JNK signaling has a profound and broad effect on activated macrophage gene expression. Our findings suggest that TNF’s anti-M2 effects evolved to specifically modulate components of tissue and reparative M2 macrophages and TNF is therefore a context-specific modulator of M2 macrophages rather than a pan-M2 inhibitor.
Collapse
Affiliation(s)
| | - David E Sanin
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany.,The Bloomberg∼Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Carolin K Koss
- Boehringer Ingelheim Pharma GmbH and Co KG, Biberach, Germany
| | | | - Andreas Petzold
- Deep Sequencing Group, Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Maria C Tanzer
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Andreas Dahl
- Deep Sequencing Group, Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Agnieszka M Kabat
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany.,The Bloomberg∼Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | | | - Leonie Zeitler
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | | | - Matthias Mann
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Axel Roers
- Institute for Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Sabine A Eming
- Department of Dermatology, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.,Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | | | - Edward J Pearce
- Department of Immunometabolism, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany.,The Bloomberg∼Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
15
|
Chen F, El-Naccache DW, Ponessa JJ, Lemenze A, Espinosa V, Wu W, Lothstein K, Jin L, Antao O, Weinstein JS, Damani-Yokota P, Khanna K, Murray PJ, Rivera A, Siracusa MC, Gause WC. Helminth resistance is mediated by differential activation of recruited monocyte-derived alveolar macrophages and arginine depletion. Cell Rep 2022; 38:110215. [PMID: 35021079 PMCID: PMC9403845 DOI: 10.1016/j.celrep.2021.110215] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022] Open
Abstract
Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.
Collapse
Affiliation(s)
- Fei Chen
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Darine W El-Naccache
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - John J Ponessa
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Alexander Lemenze
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Pathology, Immunology, and Laboratory Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Vanessa Espinosa
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Pediatrics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Wenhui Wu
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Katherine Lothstein
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Linhua Jin
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Olivia Antao
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Jason S Weinstein
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Payal Damani-Yokota
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Kamal Khanna
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA; Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Amariliz Rivera
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Pediatrics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Mark C Siracusa
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA.
| | - William C Gause
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA; Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|
16
|
De Sanctis F, Lamolinara A, Boschi F, Musiu C, Caligola S, Trovato R, Fiore A, Frusteri C, Anselmi C, Poffe O, Cestari T, Canè S, Sartoris S, Giugno R, Del Rosario G, Zappacosta B, Del Pizzo F, Fassan M, Dugnani E, Piemonti L, Bottani E, Decimo I, Paiella S, Salvia R, Lawlor RT, Corbo V, Park Y, Tuveson DA, Bassi C, Scarpa A, Iezzi M, Ugel S, Bronte V. Interrupting the nitrosative stress fuels tumor-specific cytotoxic T lymphocytes in pancreatic cancer. J Immunother Cancer 2022; 10:e003549. [PMID: 35022194 PMCID: PMC8756272 DOI: 10.1136/jitc-2021-003549] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest tumors owing to its robust desmoplasia, low immunogenicity, and recruitment of cancer-conditioned, immunoregulatory myeloid cells. These features strongly limit the success of immunotherapy as a single agent, thereby suggesting the need for the development of a multitargeted approach. The goal is to foster T lymphocyte infiltration within the tumor landscape and neutralize cancer-triggered immune suppression, to enhance the therapeutic effectiveness of immune-based treatments, such as anticancer adoptive cell therapy (ACT). METHODS We examined the contribution of immunosuppressive myeloid cells expressing arginase 1 and nitric oxide synthase 2 in building up a reactive nitrogen species (RNS)-dependent chemical barrier and shaping the PDAC immune landscape. We examined the impact of pharmacological RNS interference on overcoming the recruitment and immunosuppressive activity of tumor-expanded myeloid cells, which render pancreatic cancers resistant to immunotherapy. RESULTS PDAC progression is marked by a stepwise infiltration of myeloid cells, which enforces a highly immunosuppressive microenvironment through the uncontrolled metabolism of L-arginine by arginase 1 and inducible nitric oxide synthase activity, resulting in the production of large amounts of reactive oxygen and nitrogen species. The extensive accumulation of myeloid suppressing cells and nitrated tyrosines (nitrotyrosine, N-Ty) establishes an RNS-dependent chemical barrier that impairs tumor infiltration by T lymphocytes and restricts the efficacy of adoptive immunotherapy. A pharmacological treatment with AT38 ([3-(aminocarbonyl)furoxan-4-yl]methyl salicylate) reprograms the tumor microenvironment from protumoral to antitumoral, which supports T lymphocyte entrance within the tumor core and aids the efficacy of ACT with telomerase-specific cytotoxic T lymphocytes. CONCLUSIONS Tumor microenvironment reprogramming by ablating aberrant RNS production bypasses the current limits of immunotherapy in PDAC by overcoming immune resistance.
Collapse
Affiliation(s)
- Francesco De Sanctis
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Alessia Lamolinara
- Department of Neurosciences, Imaging and Clinical Sciences, Center for Advanced Studies and Technnology (CAST), G. d'Annunzio University of Chieti Pescara, Chieti, Italy
| | - Federico Boschi
- Department of Computer Science, University of Verona, Verona, Italy
| | - Chiara Musiu
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Simone Caligola
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Rosalinda Trovato
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Alessandra Fiore
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Cristina Frusteri
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Cristina Anselmi
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Ornella Poffe
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Tiziana Cestari
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Stefania Canè
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Silvia Sartoris
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, Verona, Italy
| | | | | | - Francesco Del Pizzo
- Department of Neurosciences, Imaging and Clinical Sciences, Center for Advanced Studies and Technnology (CAST), G. d'Annunzio University of Chieti Pescara, Chieti, Italy
| | - Matteo Fassan
- Department of Medicine, University of Padua, Padova, Italy
- Veneto Institute of Oncology-Institute for Hospitalization and Care Scientific, Padova, Italy
| | - Erica Dugnani
- Diabetes Research Institute, San Raffaele Research Centre, Milano, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, San Raffaele Research Centre, Milano, Italy
- School of Medicine and Surgery, Vita-Salute San Raffaele University, Milano, Italy
| | - Emanuela Bottani
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| | - Ilaria Decimo
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| | - Salvatore Paiella
- General and Pancreatic Surgery Unit, University of Verona, Verona, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery Unit, University of Verona, Verona, Italy
| | | | - Vincenzo Corbo
- ARC-NET, University of Verona, Verona, Italy
- Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
- Pancreatic Cancer Research Laboratory, Lustgarten Foundation, Cold Spring Harbor, New York, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
- Pancreatic Cancer Research Laboratory, Lustgarten Foundation, Cold Spring Harbor, New York, USA
| | - Claudio Bassi
- General and Pancreatic Surgery Unit, University of Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-NET, University of Verona, Verona, Italy
- Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Manuela Iezzi
- Department of Neurosciences, Imaging and Clinical Sciences, Center for Advanced Studies and Technnology (CAST), G. d'Annunzio University of Chieti Pescara, Chieti, Italy
| | - Stefano Ugel
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Vincenzo Bronte
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| |
Collapse
|
17
|
Miyamoto T, Murakami R, Hamanishi J, Tanigaki K, Hosoe Y, Mise N, Takamatsu S, Mise Y, Ukita M, Taki M, Yamanoi K, Horikawa N, Abiko K, Yamaguchi K, Baba T, Matsumura N, Mandai M. B7-H3 Suppresses Antitumor Immunity via the CCL2-CCR2-M2 Macrophage Axis and Contributes to Ovarian Cancer Progression. Cancer Immunol Res 2022; 10:56-69. [PMID: 34799346 PMCID: PMC9414298 DOI: 10.1158/2326-6066.cir-21-0407] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/03/2021] [Accepted: 11/18/2021] [Indexed: 01/07/2023]
Abstract
New approaches beyond PD-1/PD-L1 inhibition are required to target the immunologically diverse tumor microenvironment (TME) in high-grade serous ovarian cancer (HGSOC). In this study, we explored the immunosuppressive effect of B7-H3 (CD276) via the CCL2-CCR2-M2 macrophage axis and its potential as a therapeutic target. Transcriptome analysis revealed that B7-H3 is highly expressed in PD-L1-low, nonimmunoreactive HGSOC tumors, and its expression negatively correlated with an IFNγ signature, which reflects the tumor immune reactivity. In syngeneic mouse models, B7-H3 (Cd276) knockout (KO) in tumor cells, but not in stromal cells, suppressed tumor progression, with a reduced number of M2 macrophages and an increased number of IFNγ+CD8+ T cells. CCL2 expression was downregulated in the B7-H3 KO tumor cell lines. Inhibition of the CCL2-CCR2 axis partly negated the effects of B7-H3 suppression on M2 macrophage migration and differentiation, and tumor progression. In patients with HGSOC, B7-H3 expression positively correlated with CCL2 expression and M2 macrophage abundance, and patients with B7-H3-high tumors had fewer tumoral IFNγ+CD8+ T cells and poorer prognosis than patients with B7-H3-low tumors. Thus, B7-H3 expression in tumor cells contributes to CCL2-CCR2-M2 macrophage axis-mediated immunosuppression and tumor progression. These findings provide new insights into the immunologic TME and could aid the development of new therapeutic approaches against the unfavorable HGSOC phenotype.
Collapse
Affiliation(s)
- Taito Miyamoto
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Gynecology, Shiga General Hospital, Moriyama, Japan.,Corresponding Author: Ryusuke Murakami, Department of Gynecology, Shiga General Hospital, 5-4-30, Moriyama, Moriyama City, Shiga 524-8524, Japan. Phone: 817-7582-5031; Fax: 817-7582-5931; E-mail:
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Yuko Hosoe
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nathan Mise
- Department of Environmental and Preventive Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Shiro Takamatsu
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuka Mise
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayo Ukita
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoki Horikawa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Obstetrics and Gynecology, Shizuoka General Hospital, Shizuoka, Japan
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University School of Medicine, Higashiosaka, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Wan M, Ding Y, Li Z, Wang X, Xu M. Metabolic manipulation of the tumour immune microenvironment. Immunology 2021; 165:290-300. [PMID: 34962655 DOI: 10.1111/imm.13444] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/26/2022] Open
Abstract
In the past few years, the evolution of immunotherapy has resulted in a shift in cancer treatment models. However, with immunosuppressive effects of the tumour microenvironment continue to limit advances in tumour immunotherapy. The tumour microenvironment induces metabolic reprogramming in cancer cells, which results in competition for nutrients between tumour cells and host immunocytes. Metabolic and waste products originating in tumour cells can influence the activation and effector properties of immunocytes in numerous ways and ultimately promote the survival and propagation of tumour cells. In this paper, we discuss metabolic reprogramming in tumour cells and the influence of metabolite byproducts on the immune microenvironment, providing novel insights into tumour immunotherapy.
Collapse
Affiliation(s)
- Mengtian Wan
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Yuzhu Ding
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Zheng Li
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Xu Wang
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| |
Collapse
|
19
|
Moreira C, Hétru J, Paiola M, Duflot A, Chan P, Vaudry D, Pinto PIS, Monsinjon T, Knigge T. Proteomic changes in the extracellular environment of sea bass thymocytes exposed to 17α-ethinylestradiol in vitro. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 40:100911. [PMID: 34583305 DOI: 10.1016/j.cbd.2021.100911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 08/14/2021] [Accepted: 08/28/2021] [Indexed: 11/28/2022]
Abstract
The thymus is an important immune organ providing the necessary microenvironment for the development of a diverse, self-tolerant T cell repertoire, which is selected to allow for the recognition of foreign antigens while avoiding self-reactivity. Thymus function and activity are known to be regulated by sex steroid hormones, such as oestrogen, leading to sexual dimorphisms in immunocompetence between males and females. The oestrogenic modulation of the thymic function provides a potential target for environmental oestrogens, such as 17α-ethynylestradiol (EE2), to interfere with the cross-talk between the endocrine and the immune system. Oestrogen receptors have been identified on thymocytes and the thymic microenvironment, but it is unclear how oestrogens regulate thymic epithelial and T cell communication including paracrine signalling. Much less is known regarding intrathymic signalling in fish. Secretomics allows for the analysis of complex mixtures of immunomodulatory signalling factors secreted by T cells. Thus, in the present study, isolated thymocytes of the European sea bass, Dicentrarchus labrax, were exposed in vitro to 30 nM EE2 for 4 h and the T cell-secretome (i.e., extracellular proteome) was analysed by quantitative label-free mass-spectrometry. Progenesis revealed a total of 111 proteins differentially displayed between EE2-treated and control thymocytes at an α-level of 5% and a 1.3-fold change cut off (n = 5-6). The EE2-treatment significantly decreased the level of 90 proteins. Gene ontology revealed the proteasome to be the most impacted pathway. In contrast, the abundance of 21 proteins was significantly increased, with cathepsins showing the highest level of induction. However, no particular molecular pathway was significantly altered for these upregulated proteins. To the best of our knowledge, this work represents the first study of the secretome of the fish thymus exposed to the environmental oestrogen EE2, highlighting the impact on putative signalling pathways linked to immune surveillance, which may be of crucial importance for fish health and defence against pathogens.
Collapse
Affiliation(s)
- Catarina Moreira
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France
| | - Julie Hétru
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France
| | - Matthieu Paiola
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France; Department of Microbiology and Immunology, University of Rochester Medical Center, 14642 Rochester, NY, United States
| | - Aurélie Duflot
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France
| | - Philippe Chan
- Normandie Univ, UNIROUEN, PISSARO Proteomic Facility, IRIB, F-76820 Mont-Saint-Aignan, France; Normandie Univ, UNIROUEN, Institute for Research and Innovation in Biomedicine (IRIB), F-76183 Rouen, France
| | - David Vaudry
- Normandie Univ, UNIROUEN, PISSARO Proteomic Facility, IRIB, F-76820 Mont-Saint-Aignan, France; Normandie Univ, UNIROUEN, Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Inserm U1239, 76821 Mont-Saint-Aignan, France; Normandie Univ, UNIROUEN, Institute for Research and Innovation in Biomedicine (IRIB), F-76183 Rouen, France
| | - Patrícia I S Pinto
- Centro de Ciências Do Mar (CCMAR), Universidade Do Algarve, 8005-139 Faro, Portugal
| | - Tiphaine Monsinjon
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France
| | - Thomas Knigge
- Normandie Univ, UNILEHAVRE, FR CNRS 3730 SCALE, UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), F-76600 Le Havre, France.
| |
Collapse
|
20
|
Metabolic orchestration of the wound healing response. Cell Metab 2021; 33:1726-1743. [PMID: 34384520 DOI: 10.1016/j.cmet.2021.07.017] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/16/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Wound healing requires cooperation between different cell types, among which macrophages play a central role. In particular, inflammatory macrophages are engaged in the initial response to wounding, and alternatively activated macrophages are essential for wound closure and the resolution of tissue repair. The links between temporal activation-induced changes in the metabolism of such macrophages and the influence this has on their functional states, along with the realization that metabolites play both intrinsic and extrinsic roles in the cells that produce them, has focused attention on the metabolism of wound healing. Here, we discuss macrophage metabolism during distinct stages of normal healing and its related pathologic processes, such as during cancer and fibrosis. Further, we frame these insights in a broader context of the current understanding of macrophage metabolic reprogramming linked to cellular activation and function. Finally, we discuss parallels between the metabolism of macrophages and fibroblasts, the latter being a key stromal cell type in wound healing, and consider the importance of the metabolic interplay between different cell types in the wound microenvironment.
Collapse
|
21
|
An P, Xu W, Luo J, Luo Y. Expanding TOR Complex 2 Signaling: Emerging Regulators and New Connections. Front Cell Dev Biol 2021; 9:713806. [PMID: 34395443 PMCID: PMC8363310 DOI: 10.3389/fcell.2021.713806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022] Open
Abstract
Almost three decades after its seminal discovery, our understanding of the remarkable TOR pathway continues to expand. As a TOR complex, TORC2 lies at the nexus of many signaling pathways and directs a diverse array of fundamental processes such as cell survival, proliferation, and metabolism by integrating environmental and intracellular cues. The dysregulation of TORC2 activity disrupts cellular homeostasis and leads to many pathophysiological conditions. With continued efforts at mapping the signaling landscape, the pace of discovery in TORC2 regulation has been accelerated in recent years. Consequently, emerging evidence has expanded the repertoire of upstream regulators and has revealed unexpected diversity in the modes of TORC2 regulation. Multiple environmental cues and plasma membrane proteins that fine-tune TORC2 activity are unfolding. Furthermore, TORC2 signaling is intricately intertwined with other major signaling pathways. Therefore, feedback and crosstalk regulation also extensively modulate TORC2. In this context, we provide a comprehensive overview of revolutionary concepts regarding emerging regulators of TORC2 and discuss evidence of feedback and crosstalk regulation that shed new light on TORC2 biology.
Collapse
Affiliation(s)
| | | | - Junjie Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yongting Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
22
|
Van de Velde LA, Allen EK, Crawford JC, Wilson TL, Guy CS, Russier M, Zeitler L, Bahrami A, Finkelstein D, Pelletier S, Schultz-Cherry S, Thomas PG, Murray PJ. Neuroblastoma Formation Requires Unconventional CD4 T Cells and Arginase-1-Dependent Myeloid Cells. Cancer Res 2021; 81:5047-5059. [PMID: 34301764 PMCID: PMC8488023 DOI: 10.1158/0008-5472.can-21-0691] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/29/2021] [Accepted: 07/22/2021] [Indexed: 01/07/2023]
Abstract
Immune cells regulate tumor growth by mirroring their function as tissue repair organizers in normal tissues. To understand the different facets of immune-tumor collaboration through genetics, spatial transcriptomics, and immunologic manipulation with noninvasive, longitudinal imaging, we generated a penetrant double oncogene-driven autochthonous model of neuroblastoma. Spatial transcriptomic analysis showed that CD4+ and myeloid populations colocalized within the tumor parenchyma, while CD8+ T cells and B cells were peripherally dispersed. Depletion of CD4+ T cells or CCR2+ macrophages, but not B cells, CD8+ T cells, or natural killer (NK) cells, prevented tumor formation. Tumor CD4+ T cells displayed unconventional phenotypes and were clonotypically diverse and antigen independent. Within the myeloid fraction, tumor growth required myeloid cells expressing arginase-1. Overall, these results demonstrate how arginine-metabolizing myeloid cells conspire with pathogenic CD4+ T cells to create permissive conditions for tumor formation, suggesting that these protumorigenic pathways could be disabled by targeting myeloid arginine metabolism. SIGNIFICANCE: A new model of human neuroblastoma provides ways to track tumor formation and expansion in living animals, allowing identification of CD4+ T-cell and macrophage functions required for oncogenesis.
Collapse
Affiliation(s)
- Lee-Ann Van de Velde
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - E. Kaitlynn Allen
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Taylor L. Wilson
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Clifford S. Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Marion Russier
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Leonie Zeitler
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Armita Bahrami
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stephane Pelletier
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Corresponding Authors: Peter J. Murray, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried 82152, Germany. Phone: 49-89-8578-2428; E-mail: ; and Paul G. Thomas, Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105. Phone: 901-595-6507; E-mail:
| | - Peter J. Murray
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Max Planck Institute of Biochemistry, Martinsried, Germany.,Institute of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technical University of Munich, München, Germany.,Corresponding Authors: Peter J. Murray, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried 82152, Germany. Phone: 49-89-8578-2428; E-mail: ; and Paul G. Thomas, Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105. Phone: 901-595-6507; E-mail:
| |
Collapse
|
23
|
Extracellular vesicles in immunomodulation and tumor progression. Nat Immunol 2021; 22:560-570. [PMID: 33753940 PMCID: PMC9389600 DOI: 10.1038/s41590-021-00899-0] [Citation(s) in RCA: 339] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023]
Abstract
Extracellular vesicles have emerged as prominent regulators of the immune response during tumor progression. EVs contain a diverse repertoire of molecular cargo that plays a critical role in immunomodulation. Here, we identify the role of EVs as mediators of communication between cancer and immune cells. This expanded role of EVs may shed light on the mechanisms behind tumor progression and provide translational diagnostic and prognostic tools for immunologists.
Collapse
|
24
|
Badeaux MD, Rolig AS, Agnello G, Enzler D, Kasiewicz MJ, Priddy L, Wiggins JF, Muir A, Sullivan MR, Van Cleef J, Daige C, Vander Heiden MG, Rajamanickam V, Wooldridge JE, Redmond WL, Rowlinson SW. Arginase Therapy Combines Effectively with Immune Checkpoint Blockade or Agonist Anti-OX40 Immunotherapy to Control Tumor Growth. Cancer Immunol Res 2021; 9:415-429. [PMID: 33500272 DOI: 10.1158/2326-6066.cir-20-0317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/18/2020] [Accepted: 01/22/2021] [Indexed: 11/16/2022]
Abstract
Metabolic dysregulation is a hallmark of cancer. Many tumors exhibit auxotrophy for various amino acids, such as arginine, because they are unable to meet the demand for these amino acids through endogenous production. This vulnerability can be exploited by employing therapeutic strategies that deplete systemic arginine in order to limit the growth and survival of arginine auxotrophic tumors. Pegzilarginase, a human arginase-1 enzyme engineered to have superior stability and enzymatic activity relative to the native human arginase-1 enzyme, depletes systemic arginine by converting it to ornithine and urea. Therapeutic administration of pegzilarginase in the setting of arginine auxotrophic tumors exerts direct antitumor activity by starving the tumor of exogenous arginine. We hypothesized that in addition to this direct effect, pegzilarginase treatment indirectly augments antitumor immunity through increased antigen presentation, thus making pegzilarginase a prime candidate for combination therapy with immuno-oncology (I-O) agents. Tumor-bearing mice (CT26, MC38, and MCA-205) receiving pegzilarginase in combination with anti-PD-L1 or agonist anti-OX40 experienced significantly increased survival relative to animals receiving I-O monotherapy. Combination pegzilarginase/immunotherapy induced robust antitumor immunity characterized by increased intratumoral effector CD8+ T cells and M1 polarization of tumor-associated macrophages. Our data suggest potential mechanisms of synergy between pegzilarginase and I-O agents that include increased intratumoral MHC expression on both antigen-presenting cells and tumor cells, and increased presence of M1-like antitumor macrophages. These data support the clinical evaluation of I-O agents in conjunction with pegzilarginase for the treatment of patients with cancer.
Collapse
Affiliation(s)
| | - Annah S Rolig
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | | | | | - Melissa J Kasiewicz
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | | | | | - Alexander Muir
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois
| | - Mark R Sullivan
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | | | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon.
| | | |
Collapse
|
25
|
Zhang J, Yi C, Han J, Ming T, Zhou J, Lu C, Li Y, Su X. Novel high-docosahexaenoic-acid tuna oil supplementation modulates gut microbiota and alleviates obesity in high-fat diet mice. Food Sci Nutr 2020; 8:6513-6527. [PMID: 33312536 PMCID: PMC7723182 DOI: 10.1002/fsn3.1941] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/08/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
Studies have documented the benefits of fish oil in different diseases because of its high n-3 polyunsaturated fatty acid content. However, these studies mostly used commercially available fish oil supplements with a ratio of 18/12 for eicosapentaenoic acid and docosahexaenoic acid (DHA). However, increasing DHA content for this commonly used ratio might bring out a varied metabolic effect, which have remained unclear. Thus, in this study, a novel tuna oil (TO) was applied to investigate the effect of high-DHA content on the alteration of the gut microbiota and obesity in high-fat diet mice. The results suggest that high-DHA TO (HDTO) supplementation notably ameliorates obesity and related lipid parameters and restores the expression of lipid metabolism-related genes. The benefits of TOs were derived from their modification of the gut microbiota composition and structure in mice. A high-fat diet triggered an increased Firmicutes/Bacteroidetes ratio that was remarkably restored by TOs. The number of obesity-promoting bacteria-Desulfovibrio, Paraeggerthella, Terrisporobacter, Millionella, Lachnoclostridium, Anaerobacterium, and Ruminiclostridium-was dramatically reduced. Desulfovibrio desulfuricans, Alistipes putredinis, and Millionella massiliensis, three dysbiosis-related species, were especially regulated by HDTO. Regarding the prevention of obesity, HDTO outperforms the normal TO. Intriguingly, HDTO feeding to HFD-fed mice might alter the arginine and proline metabolism of intestinal microbiota.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory for Quality and Safety of Argo‐productsNingbo UniversityNingboChina
- School of Marine ScienceNingbo UniversityNingboChina
- Faculty of Food ScienceZhejiang Pharmaceutical CollegeNingboChina
| | - Congmin Yi
- State Key Laboratory for Quality and Safety of Argo‐productsNingbo UniversityNingboChina
- School of Marine ScienceNingbo UniversityNingboChina
| | - Jiaojiao Han
- State Key Laboratory for Quality and Safety of Argo‐productsNingbo UniversityNingboChina
- School of Marine ScienceNingbo UniversityNingboChina
| | - Tinghong Ming
- State Key Laboratory for Quality and Safety of Argo‐productsNingbo UniversityNingboChina
- School of Marine ScienceNingbo UniversityNingboChina
| | - Jun Zhou
- State Key Laboratory for Quality and Safety of Argo‐productsNingbo UniversityNingboChina
- School of Marine ScienceNingbo UniversityNingboChina
| | - Chenyang Lu
- State Key Laboratory for Quality and Safety of Argo‐productsNingbo UniversityNingboChina
- School of Marine ScienceNingbo UniversityNingboChina
| | - Ye Li
- State Key Laboratory for Quality and Safety of Argo‐productsNingbo UniversityNingboChina
- School of Marine ScienceNingbo UniversityNingboChina
| | - Xiurong Su
- State Key Laboratory for Quality and Safety of Argo‐productsNingbo UniversityNingboChina
- School of Marine ScienceNingbo UniversityNingboChina
| |
Collapse
|
26
|
Preparation and purification of an immunoregulatory peptide from Stolephorus chinensis of the East Sea of China. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.08.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Lercher A, Popa AM, Viczenczova C, Kosack L, Klavins K, Agerer B, Opitz CA, Lanz TV, Platten M, Bergthaler A. Hepatocyte-intrinsic type I interferon signaling reprograms metabolism and reveals a novel compensatory mechanism of the tryptophan-kynurenine pathway in viral hepatitis. PLoS Pathog 2020; 16:e1008973. [PMID: 33045014 PMCID: PMC7580883 DOI: 10.1371/journal.ppat.1008973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/22/2020] [Accepted: 09/10/2020] [Indexed: 12/27/2022] Open
Abstract
The liver is a central regulator of metabolic homeostasis and serum metabolite levels. Hepatocytes are the functional units of the liver parenchyma and not only responsible for turnover of biomolecules but also act as central immune signaling platforms. Hepatotropic viruses infect liver tissue, resulting in inflammatory responses, tissue damage and hepatitis. Combining well-established in vitro and in vivo model systems with transcriptomic analyses, we show that type I interferon signaling initiates a robust antiviral immune response in hepatocytes. Strikingly, we also identify IFN-I as both, sufficient and necessary, to induce wide-spread metabolic reprogramming in hepatocytes. IFN-I specifically rewired tryptophan metabolism and induced hepatic tryptophan oxidation to kynurenine via Tdo2, correlating with altered concentrations of serum metabolites upon viral infection. Infected Tdo2-deficient animals displayed elevated serum levels of tryptophan and, unexpectedly, also vast increases in the downstream immune-suppressive metabolite kynurenine. Thus, Tdo2-deficiency did not result in altered serum homeostasis of the tryptophan to kynurenine ratio during infection, which seemed to be independent of hepatocyte-intrinsic compensation via the IDO-axis. These data highlight that inflammation-induced reprogramming of systemic tryptophan metabolism is tightly regulated in viral hepatitis. Viral hepatitis is responsible for more than one million annual deaths worldwide and may progress to liver cirrhosis and hepatocellular carcinoma. The main metabolic cell type of the liver is the hepatocyte. In viral hepatitis, type I interferon (IFN-I) signaling rewires hepatocyte metabolism and serum metabolites to shape disease pathophysiology – an immune-regulatory circuit that might be therapeutically exploited. Here, we show that hepatocyte-intrinsic antiviral IFN-I signaling is both necessary and sufficient to induce wide-spread metabolic changes in hepatocytes. We identify a IFN-I-mediated induction of the hepatic kynurenine pathway via the rate-limiting and liver-specific enzyme TDO2, which controls serum homeostasis of tryptophan by converting it into kynurenine. Loss of TDO2 triggers a so far unknown compensatory mechanism, resulting in a vast increase of circulating kynurenine independent of hepatocyte intrinsic activity of the related IDO-enzymes. This study provides new insights into how inflammation reprograms metabolism of the liver and the kynurenine pathway during viral hepatitis.
Collapse
MESH Headings
- Animals
- Antiviral Agents/metabolism
- Female
- Hepatitis Viruses/isolation & purification
- Hepatitis, Viral, Animal/immunology
- Hepatitis, Viral, Animal/metabolism
- Hepatitis, Viral, Animal/virology
- Hepatocytes/immunology
- Hepatocytes/metabolism
- Hepatocytes/virology
- Humans
- Immunity, Innate/immunology
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/virology
- Interferon Regulatory Factor-7/physiology
- Kynurenine/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, Interferon alpha-beta/physiology
- STAT1 Transcription Factor/physiology
- Tryptophan/metabolism
- Tryptophan Oxygenase/physiology
Collapse
Affiliation(s)
- Alexander Lercher
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
- * E-mail: (AL); (AB)
| | - Alexandra M. Popa
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Csilla Viczenczova
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Lindsay Kosack
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Kristaps Klavins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Benedikt Agerer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
| | - Christiane A. Opitz
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurology Clinic and National Center for Tumor Diseases, University Hospital of Heidelberg, Heidelberg, Germany
| | - Tobias V. Lanz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
- Department of Neurology, University of Heidelberg, Medical Faculty Mannheim, Mannheim, Germany
| | - Michael Platten
- Department of Neurology, University of Heidelberg, Medical Faculty Mannheim, Mannheim, Germany
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse, Vienna, Austria
- * E-mail: (AL); (AB)
| |
Collapse
|
28
|
Borek B, Gajda T, Golebiowski A, Blaszczyk R. Boronic acid-based arginase inhibitors in cancer immunotherapy. Bioorg Med Chem 2020; 28:115658. [PMID: 32828425 DOI: 10.1016/j.bmc.2020.115658] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/27/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023]
Abstract
Arginase is an enzyme that converts l-arginine to l-ornithine and urea in the urea cycle. There are two isoforms of arginase in mammals: ARG-1 and ARG-2. l-Arginine level changes occur in patients with various types of affliction. An overexpression of arginase leads to the depletion of arginine and then to inhibition of the growth of T and NK cells, and in effect to the tumor escape of the immune response. Based on those observations, an inhibition of arginase is proposed as a method to improve anti-tumor immune responses (via an activation and proliferation of T and NK cells). Boronic acid derivatives as arginase inhibitors are leading, potential therapeutic agents for the treatment of several diseases. All these compounds are derived from the original 2-(S)-amino-6-boronohexanoic acid (ABH), the first boronic acid arginase inhibitor proposed by Christianson et al. This article focuses on the review of such sub-class of arginase inhibitors and highlights their SAR and PK properties. It covers molecules published until early 2020, including patent applications.
Collapse
Affiliation(s)
- Bartlomiej Borek
- OncoArendi Therapeutics SA, 101 Żwirki i Wigury St, 02-089 Warsaw, Poland.
| | - Tadeusz Gajda
- Institute of Organic Chemistry, Faculty of Chemistry, Technical University of Lodz, 116 Stefana Żeromskiego St, 90-924 Łódź, Poland
| | - Adam Golebiowski
- OncoArendi Therapeutics SA, 101 Żwirki i Wigury St, 02-089 Warsaw, Poland
| | - Roman Blaszczyk
- OncoArendi Therapeutics SA, 101 Żwirki i Wigury St, 02-089 Warsaw, Poland
| |
Collapse
|
29
|
Huang H, Long L, Zhou P, Chapman NM, Chi H. mTOR signaling at the crossroads of environmental signals and T-cell fate decisions. Immunol Rev 2020; 295:15-38. [PMID: 32212344 PMCID: PMC8101438 DOI: 10.1111/imr.12845] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/19/2020] [Indexed: 12/28/2022]
Abstract
The evolutionarily conserved serine/threonine kinase mTOR (mechanistic target of rapamycin) forms the distinct protein complexes mTORC1 and mTORC2 and integrates signals from the environment to coordinate downstream signaling events and various cellular processes. T cells rely on mTOR activity for their development and to establish their homeostasis and functional fitness. Here, we review recent progress in our understanding of the upstream signaling and downstream targets of mTOR. We also provide an updated overview of the roles of mTOR in T-cell development, homeostasis, activation, and effector-cell fate decisions, as well as its important impacts on the suppressive activity of regulatory T cells. Moreover, we summarize the emerging roles of mTOR in T-cell exhaustion and transdifferentiation. A better understanding of the contribution of mTOR to T-cell fate decisions will ultimately aid in the therapeutic targeting of mTOR in human disease.
Collapse
Affiliation(s)
- Hongling Huang
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Lingyun Long
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Equal contribution
| | - Peipei Zhou
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Equal contribution
| | - Nicole M. Chapman
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
30
|
Aminoacyl-tRNA synthetase inhibition activates a pathway that branches from the canonical amino acid response in mammalian cells. Proc Natl Acad Sci U S A 2020; 117:8900-8911. [PMID: 32253314 DOI: 10.1073/pnas.1913788117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Signaling pathways that sense amino acid abundance are integral to tissue homeostasis and cellular defense. Our laboratory has previously shown that halofuginone (HF) inhibits the prolyl-tRNA synthetase catalytic activity of glutamyl-prolyl-tRNA synthetase (EPRS), thereby activating the amino acid response (AAR). We now show that HF treatment selectively inhibits inflammatory responses in diverse cell types and that these therapeutic benefits occur in cells that lack GCN2, the signature effector of the AAR. Depletion of arginine, histidine, or lysine from cultured fibroblast-like synoviocytes recapitulates key aspects of HF treatment, without utilizing GCN2 or mammalian target of rapamycin complex 1 pathway signaling. Like HF, the threonyl-tRNA synthetase inhibitor borrelidin suppresses the induction of tissue remodeling and inflammatory mediators in cytokine-stimulated fibroblast-like synoviocytes without GCN2, but both aminoacyl-tRNA synthetase (aaRS) inhibitors are sensitive to the removal of GCN1. GCN1, an upstream component of the AAR pathway, binds to ribosomes and is required for GCN2 activation. These observations indicate that aaRS inhibitors, like HF, can modulate inflammatory response without the AAR/GCN2 signaling cassette, and that GCN1 has a role that is distinct from its activation of GCN2. We propose that GCN1 participates in a previously unrecognized amino acid sensor pathway that branches from the canonical AAR.
Collapse
|
31
|
Munshi S, Loh MK, Ferrara N, DeJoseph MR, Ritger A, Padival M, Record MJ, Urban JH, Rosenkranz JA. Repeated stress induces a pro-inflammatory state, increases amygdala neuronal and microglial activation, and causes anxiety in adult male rats. Brain Behav Immun 2020; 84:180-199. [PMID: 31785394 PMCID: PMC7010555 DOI: 10.1016/j.bbi.2019.11.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/25/2022] Open
Abstract
A link exists between immune function and psychiatric conditions, particularly depressive and anxiety disorders. Psychological stress is a powerful trigger for these disorders and stress influences immune state. However, the nature of peripheral immune changes after stress conflicts across studies, perhaps due to the focus on few measures of pro-inflammatory or anti-inflammatory processes. The basolateral amygdala (BLA) is critical for emotion, and plays an important role in the effects of stress on anxiety. As such, it may be a primary central nervous system (CNS) mediator for the effects of peripheral immune changes on anxiety after stress. Therefore, this study aimed to delineate the influence of stress on peripheral pro-inflammatory and anti-inflammatory aspects, BLA immune activation, and its impact on BLA neuronal activity. To produce a more encompassing view of peripheral immune changes, this study used a less restrictive approach to categorize and group peripheral immune changes. We found that repeated social defeat stress in adult male Sprague-Dawley rats increased the frequencies of mature T-cells positive for intracellular type 2-like cytokine and serum pro-inflammatory cytokines. Principal component analysis and hierarchical clustering was used to guide grouping of T-cells and cytokines, producing unique profiles. Stress shifted the balance towards a specific set that included mostly type 2-like T-cells and pro-inflammatory cytokines. Within the CNS component, repeated stress caused an increase of activated microglia in the BLA, increased anxiety-like behaviors across several assays, and increased BLA neuronal firing in vivo that was prevented by blockade of microglia activation. Because repeated stress can trigger anxiety states by actions in the BLA, and altered immune function can trigger anxiety, these results suggest that repeated stress may trigger anxiety-like behaviors by inducing a pro-inflammatory state in the periphery and the BLA. These results begin to uncover how stress may recruit the immune system to alter the function of brain regions critical to emotion.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Department of Foundational Sciences and Humanities, Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Maxine K. Loh
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Nicole Ferrara
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - M. Regina DeJoseph
- Department of Foundational Sciences and Humanities, Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Alexandra Ritger
- Department of Foundational Sciences and Humanities, Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Mallika Padival
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Matthew J. Record
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Janice H. Urban
- Department of Foundational Sciences and Humanities, Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - J. Amiel Rosenkranz
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Corresponding Author: J. Amiel Rosenkranz, Ph.D., Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA., Telephone: 847-578-8680; Fax: 847-578-3268,
| |
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: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Fu L, Fu S, Wang C, Xie M, Wang Y. Yogurt-sourced probiotic bacteria alleviate shrimp tropomyosin-induced allergic mucosal disorders, potentially through microbiota and metabolism modifications. Allergol Int 2019; 68:506-514. [PMID: 31262632 DOI: 10.1016/j.alit.2019.05.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/19/2019] [Accepted: 05/11/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Shrimp tropomyosin (TM) is a major food allergen that may cause serious allergic responses, lactic acid-producing bacteria (LAPB) are believed to alleviate food allergy, but the mechanisms have not been fully clarified. The aim of this work is to investigate the mechanisms of LAPB in ameliorating food allergy-induced intestinal mucosal disorders and to investigate whether or not these disorders occur by the regulation of gut microbiota and metabolism. METHODS A TM allergy BALB/c mouse model was established, and two LAPB strains, Bifidobacterium longum (Bi) and Bacillus coagulans (Bc), were used for oral treatment in sensitized mice. The allergic mucosal disorders were assessed by histological analysis and ELISAs. Additionally, microbiota and metabolic modifications were determined by 16S rRNA gene amplicon sequencing and GC-TOF-MS, respectively. RESULTS YSPB administration suppressed TM-induced intestinal mucosal disorders, restored allergenic Th2 cell over-polarization and dysbiosis, and regulated gut arginine and proline metabolism pathways. Statistical analysis suggested the metabolites aspartate and arginine, as well as several commensal flora groups, to be the critical mediators in the process. CONCLUSIONS These data demonstrated the correlation between allergic mucosal disorder, T cell subtype differentiation, gut microbiota composition and intestinal metabolism especially the arginine and proline metabolism pathways. We also revealed the significant effects of LAPB in ameliorating food allergy and maintaining the mucosal ecosystem. This study confirmed the efficiency of LAPB in relieving food allergy, provided Bi and Bc as the potential treatment approaches, and suggested amino acid metabolism pathways might be the novel targets for potential clinical applications.
Collapse
Affiliation(s)
- Linglin Fu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China; Zhejiang Engineering Institute of Food Quality and Safety, Zhejiang Gongshang University, Hangzhou, China
| | - Shujie Fu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Chong Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Menghua Xie
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Yanbo Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China; Zhejiang Engineering Institute of Food Quality and Safety, Zhejiang Gongshang University, Hangzhou, China.
| |
Collapse
|
34
|
Small extracellular vesicles containing arginase-1 suppress T-cell responses and promote tumor growth in ovarian carcinoma. Nat Commun 2019; 10:3000. [PMID: 31278254 PMCID: PMC6611910 DOI: 10.1038/s41467-019-10979-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 06/12/2019] [Indexed: 12/21/2022] Open
Abstract
Tumor-driven immune suppression is a major barrier to successful immunotherapy in ovarian carcinomas (OvCa). Among various mechanisms responsible for immune suppression, arginase-1 (ARG1)-carrying small extracellular vesicles (EVs) emerge as important contributors to tumor growth and tumor escape from the host immune system. Here, we report that small EVs found in the ascites and plasma of OvCa patients contain ARG1. EVs suppress proliferation of CD4+ and CD8+ T-cells in vitro and in vivo in OvCa mouse models. In mice, ARG1-containing EVs are transported to draining lymph nodes, taken up by dendritic cells and inhibit antigen-specific T-cell proliferation. Increased expression of ARG1 in mouse OvCa cells is associated with accelerated tumor progression that can be blocked by an arginase inhibitor. Altogether, our studies show that tumor cells use EVs as vehicles to carry over long distances and deliver to immune cells a metabolic checkpoint molecule – ARG1, mitigating anti-tumor immune responses. Cancer cells employ a variety of ways to escape the immune system. Here, the authors show that ovarian cancer cells produce small extracellular vescicles containing arginase 1 that are taken up by dendritic cells in the draining lymph nodes, resulting in inhibition of antigen-specific T-cell proliferation.
Collapse
|
35
|
Wnt Signaling in Cancer Metabolism and Immunity. Cancers (Basel) 2019; 11:cancers11070904. [PMID: 31261718 PMCID: PMC6678221 DOI: 10.3390/cancers11070904] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/22/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022] Open
Abstract
The Wingless (Wnt)/β-catenin pathway has long been associated with tumorigenesis, tumor plasticity, and tumor-initiating cells called cancer stem cells (CSCs). Wnt signaling has recently been implicated in the metabolic reprogramming of cancer cells. Aberrant Wnt signaling is considered to be a driver of metabolic alterations of glycolysis, glutaminolysis, and lipogenesis, processes essential to the survival of bulk and CSC populations. Over the past decade, the Wnt pathway has also been shown to regulate the tumor microenvironment (TME) and anti-cancer immunity. Wnt ligands released by tumor cells in the TME facilitate the immune evasion of cancer cells and hamper immunotherapy. In this review, we illustrate the role of the canonical Wnt/β-catenin pathway in cancer metabolism and immunity to explore the potential therapeutic approach of targeting Wnt signaling from a metabolic and immunological perspective.
Collapse
|
36
|
Paduch K, Debus A, Rai B, Schleicher U, Bogdan C. Resolution of Cutaneous Leishmaniasis and Persistence of Leishmania major in the Absence of Arginase 1. THE JOURNAL OF IMMUNOLOGY 2019; 202:1453-1464. [PMID: 30665936 DOI: 10.4049/jimmunol.1801249] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/16/2018] [Indexed: 01/29/2023]
Abstract
Arginase (Arg) 1 is expressed by hematopoietic (e.g., macrophages) and nonhematopoietic cells (e.g., endothelial cells) and converts l-arginine into ornithine and urea. The enzyme is implicated in tissue repair but also antagonizes the production of NO by type 2 NO synthase in myeloid cells and thereby impedes the control of intracellular parasites such as Leishmania major In this study, we tested whether Arg1 is required for spontaneous healing of acute cutaneous leishmaniasis in C57BL/6 mice and for lifelong parasite persistence in draining lymph nodes (dLNs) of healed mice. In vitro, bone marrow-derived macrophages and lymphoid endothelial cells readily expressed Arg1 in response to IL-4 and/or IL-13, whereas skin or dLN fibroblasts failed to do so, even during hypoxia. In vivo, Arg1 was found in skin lesions and, to a much lower extent, also in dLNs of acutely infected C57BL/6 mice but became undetectable at both sites after healing. Deletion of Arg1 in hematopoietic and endothelial cells using Tie2Cre+/-Arg1fl/fl C57BL/6 mice abolished the expression of Arg1 in skin lesions and dLNs but did not affect development and resolution of skin lesions, parasite burden, NO production, or host cell tropism of L. major during the acute or persistent phase of infection. Similar to wild-type controls, parasites persisting in Arg1-deficient mice favored NO synthase 2-negative areas and mainly resided in myeloid cells and fibroblasts. We conclude that Arg1 expression by hematopoietic and endothelial cells is completely dispensable for clinical resolution of cutaneous leishmaniasis and for long-term persistence of L. major.
Collapse
Affiliation(s)
- Katrin Paduch
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and
| | - Andrea Debus
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and
| | - Baplu Rai
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054 Erlangen, Germany; and .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
37
|
The Adaptive Complexity of Cancer. BIOMED RESEARCH INTERNATIONAL 2019; 2018:5837235. [PMID: 30627563 PMCID: PMC6304530 DOI: 10.1155/2018/5837235] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/15/2018] [Indexed: 12/13/2022]
Abstract
Cancer treatment options are expanding to the benefit of significant segments of patients. However, their therapeutic power is not equally realized for all cancer patients due to drug toxicity and disease resistance. Overcoming these therapeutic challenges would require a better understanding of the adaptive survival mechanisms of cancer. In this respect, an integrated view of the disease as a complex adaptive system is proposed as a framework to explain the dynamic coupling between the various drivers underlying tumor growth and cancer resistance to therapy. In light of this system view of cancer, the immune system is in principal the most appropriate and naturally available therapeutic instrument that can thwart the adaptive survival mechanisms of cancer. In this respect, new cancer therapies should aim at restoring immunosurveillance by priming the induction of an effective immune response through a judicious targeting of immunosuppression, inflammation, and the tumor nutritional lifeline extended by the tumor microenvironment.
Collapse
|
38
|
Ramapriyan R, Caetano MS, Barsoumian HB, Mafra ACP, Zambalde EP, Menon H, Tsouko E, Welsh JW, Cortez MA. Altered cancer metabolism in mechanisms of immunotherapy resistance. Pharmacol Ther 2018; 195:162-171. [PMID: 30439456 DOI: 10.1016/j.pharmthera.2018.11.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many metabolic alterations, including the Warburg effect, occur in cancer cells that influence the tumor microenvironment, including switching to glycolysis from oxidative phosphorylation, using opportunistic modes of nutrient acquisition, and increasing lipid biosynthesis. The altered metabolic landscape of the tumor microenvironment can suppress the infiltration of immune cells and other functions of antitumor immunity through the production of immune-suppressive metabolites. Metabolic dysregulation in cancer cells further affects the expression of cell surface markers, which interferes with immune surveillance. Immune checkpoint therapies have revolutionized the standard of care for some patients with cancer, but disease in many others is resistant to immunotherapy. Specific metabolic pathways involved in immunotherapy resistance include PI3K-Akt-mTOR, hypoxia-inducible factor (HIF), adenosine, JAK/STAT, and Wnt/Beta-catenin. Depletion of essential amino acids such as glutamine and tryptophan and production of metabolites like kynurenine in the tumor microenvironment also blunt immune cell function. Targeted therapies against metabolic checkpoints could work in synergy with immune checkpoint therapy. This combined strategy could be refined by profiling patients' mutation status before treatment and identifying the optimal sequencing of therapies. This personalized combinatorial approach, which has yet to be explored, may well pave the way for overcoming resistance to immunotherapy.
Collapse
Affiliation(s)
- Rishab Ramapriyan
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mauricio S Caetano
- Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B Barsoumian
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ana Carolina P Mafra
- Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Erika Pereira Zambalde
- Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hari Menon
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Efrosini Tsouko
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - James W Welsh
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Departments of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
39
|
Yap GS, Gause WC. Helminth Infections Induce Tissue Tolerance Mitigating Immunopathology but Enhancing Microbial Pathogen Susceptibility. Front Immunol 2018; 9:2135. [PMID: 30386324 PMCID: PMC6198046 DOI: 10.3389/fimmu.2018.02135] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/30/2018] [Indexed: 01/17/2023] Open
Abstract
Helminths are ubiquitous and have chronically infected vertebrates throughout their evolution. As such helminths have likely exerted considerable selection pressure on our immune systems. The large size of multicellular helminths and their limited replicative capacity in the host necessarily elicits different host protective mechanisms than the immune response evoked by microbial pathogens such as bacteria, viruses and intracellular parasites. The cellular damage resulting from helminth migration through tissues is a major trigger of the type 2 and regulatory immune responses, which activates wound repair mechanisms that increases tissue tolerance to injury and resistance mechanisms that enhance resistance to further colonization with larval stages. While these wound healing and anti-inflammatory responses may be beneficial to the helminth infected host, they may also compromise the host's ability to mount protective immune responses to microbial pathogens. In this review we will first describe helminth-induced tolerance mechanisms that develop in specific organs including the lung and the intestine, and how adaptive immunity may contribute to these responses through differential activation of T cells in the secondary lymphoid organs. We will then integrate studies that have examined how the immune response is modulated in these specific tissues during coinfection of helminths with viruses, protozoa, and bacteria.
Collapse
Affiliation(s)
- George S Yap
- Department of Medicine, Center for Immunity and Inflammation, Rutgers University-New Jersey Medical School, Newark, NJ, United States
| | - William C Gause
- Department of Medicine, Center for Immunity and Inflammation, Rutgers University-New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
40
|
McSorley HJ, Chayé MAM, Smits HH. Worms: Pernicious parasites or allies against allergies? Parasite Immunol 2018; 41:e12574. [PMID: 30043455 PMCID: PMC6585781 DOI: 10.1111/pim.12574] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/13/2018] [Accepted: 07/18/2018] [Indexed: 12/14/2022]
Abstract
Type 2 immune responses are most commonly associated with allergy and helminth parasite infections. Since the discovery of Th1 and Th2 immune responses more than 30 years ago, models of both allergic disease and helminth infections have been useful in characterizing the development, effector mechanisms and pathological consequences of type 2 immune responses. The observation that some helminth infections negatively correlate with allergic and inflammatory disease led to a large field of research into parasite immunomodulation. However, it is worth noting that helminth parasites are not always benign infections, and that helminth immunomodulation can have stimulatory as well as suppressive effects on allergic responses. In this review, we will discuss how parasitic infections change host responses, the consequences for bystander immunity and how this interaction influences clinical symptoms of allergy.
Collapse
Affiliation(s)
- Henry J McSorley
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mathilde A M Chayé
- Department of Parasitology, Leiden Immunology of Parasitic Infections Group, Leiden University Medical Centre, ZA Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden Immunology of Parasitic Infections Group, Leiden University Medical Centre, ZA Leiden, The Netherlands
| |
Collapse
|
41
|
Biswas A, Shouval DS, Griffith A, Goettel JA, Field M, Kang YH, Konnikova L, Janssen E, Redhu NS, Thrasher AJ, Chatila T, Kuchroo VK, Geha RS, Notarangelo LD, Pai SY, Horwitz BH, Snapper SB. WASP-mediated regulation of anti-inflammatory macrophages is IL-10 dependent and is critical for intestinal homeostasis. Nat Commun 2018; 9:1779. [PMID: 29725003 PMCID: PMC5934380 DOI: 10.1038/s41467-018-03670-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 03/03/2018] [Indexed: 01/01/2023] Open
Abstract
Mutations in Wiskott–Aldrich syndrome protein (WASP) cause autoimmune sequelae including colitis. Yet, how WASP mediates mucosal homeostasis is not fully understood. Here we show that WASP-mediated regulation of anti-inflammatory macrophages is critical for mucosal homeostasis and immune tolerance. The generation and function of anti-inflammatory macrophages are defective in both human and mice in the absence of WASP. Expression of WASP specifically in macrophages, but not in dendritic cells, is critical for regulation of colitis development. Importantly, transfer of WT anti-inflammatory macrophages prevents the development of colitis. DOCK8-deficient macrophages phenocopy the altered macrophage properties associated with WASP deficiency. Mechanistically, we show that both WASP and DOCK8 regulates macrophage function by modulating IL-10-dependent STAT3 phosphorylation. Overall, our study indicates that anti-inflammatory macrophage function and mucosal immune tolerance require both WASP and DOCK8, and that IL-10 signalling modulates a WASP-DOCK8 complex. Deficiency in Wiskott-Aldrich syndrome protein (WASP) has been associated with autoimmune colitis, but the underlying mechanism is still unclear. Here the authors show that WASP deficiency is associated with defective WASP/DOCK8 complex formation, altered IL-10 signalling, and impaired anti-inflammatory macrophage functions.
Collapse
Affiliation(s)
- Amlan Biswas
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA.,Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Dror S Shouval
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA.,Division of Pediatric Gastroenterology and Nutrition, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, 52621, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Alexandra Griffith
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Jeremy A Goettel
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA.,Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Michael Field
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Yu Hui Kang
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA.,Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Liza Konnikova
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA.,Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Erin Janssen
- Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,Division of Immunology, Boston Children's Hospital, Boston, 1 Blackfan Circle, Massachusetts, 02115, USA
| | - Naresh Singh Redhu
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA.,Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Adrian J Thrasher
- Great Ormond Street Hospital NHS Trust, London and Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Talal Chatila
- Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,Division of Immunology, Boston Children's Hospital, Boston, 1 Blackfan Circle, Massachusetts, 02115, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, 60 Fenwood Road, Boston, Massachusetts, 02115, USA
| | - Raif S Geha
- Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,Division of Immunology, Boston Children's Hospital, Boston, 1 Blackfan Circle, Massachusetts, 02115, USA
| | - Luigi D Notarangelo
- Clinical Immunology and Microbiology, NIAID, National Institutes of Health, 10 Center Drive, MSC 1456, Bethesda, Maryland, 20892-9806, USA
| | - Sung-Yun Pai
- Division of Hematology-Oncology, Boston Children's Hospital Boston, 1 Blackfan Circle, Boston, Massachusetts, 02115, USA
| | - Bruce H Horwitz
- Department of Pediatrics, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, USA.,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA.,Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, 02115, Massachusetts, USA.,Division of Emergency Medicine, Boston Children's Hospital, Boston, 300 Longwood Avenue, Boston, Massacusetts, 02115, USA
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts, 02115, USA. .,VEO-IBD Consortium, 300 Longwood Avenue, Boston, MA, 02115, USA. .,Division of Gastroenterology, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, 75 Francis Street, Boston, Massachusetts, 02115, USA.
| |
Collapse
|
42
|
Pearl JE, Das M, Cooper AM. Immunological roulette: Luck or something more? Considering the connections between host and environment in TB. Cell Mol Immunol 2018; 15:226-232. [PMID: 29375129 DOI: 10.1038/cmi.2017.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 02/07/2023] Open
Abstract
Accurate prediction of which patient will progress from a sub-clinical Mycobacterium tuberculosis infection to active tuberculosis represents an elusive, yet critical, clinical research objective. From the individual perspective, progression can be considered to be the product of a series of unfortunate events or even a run of bad luck. Here, we identify the subtle physiological relationships that can influence the odds of progression to active TB and how this progression may reflect directed dysbiosis in a number of interrelated systems. Most infected individuals who progress to disease have apparently good immune responses, but these responses are, at times, compromised by either local or systemic environmental factors. Obvious disease promoting processes, such as tissue-damaging granulomata, usually manifest in the lung, but illness is systemic. This apparent dichotomy between local and systemic reflects a clear need to define the factors that promote progression to active disease within the context of the body as a physiological whole. We discuss aspects of the host environment that can impact expression of immunity, including the microbiome, glucocorticoid-mediated regulation, catecholamines and interaction between the gut, liver and lung. We suggest the importance of integrating precision medicine into our analyses of experimental outcomes such that apparently conflicting results are not contentious, but rather reflect the impact of these subtle relationships with our environment and microbiota.
Collapse
Affiliation(s)
- John E Pearl
- Leicester Tuberculosis Research Group (LTBRG), Department of Infection Immunity and Inflammation, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Mrinal Das
- Leicester Tuberculosis Research Group (LTBRG), Department of Infection Immunity and Inflammation, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Andrea M Cooper
- Leicester Tuberculosis Research Group (LTBRG), Department of Infection Immunity and Inflammation, University of Leicester, University Road, Leicester LE1 7RH, UK
| |
Collapse
|
43
|
Na YR, Je S, Seok SH. Metabolic features of macrophages in inflammatory diseases and cancer. Cancer Lett 2018; 413:46-58. [PMID: 29100962 DOI: 10.1016/j.canlet.2017.10.044] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/17/2017] [Accepted: 10/27/2017] [Indexed: 12/31/2022]
|
44
|
Steggerda SM, Bennett MK, Chen J, Emberley E, Huang T, Janes JR, Li W, MacKinnon AL, Makkouk A, Marguier G, Murray PJ, Neou S, Pan A, Parlati F, Rodriguez MLM, Van de Velde LA, Wang T, Works M, Zhang J, Zhang W, Gross MI. Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J Immunother Cancer 2017; 5:101. [PMID: 29254508 PMCID: PMC5735564 DOI: 10.1186/s40425-017-0308-4] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/30/2017] [Indexed: 02/08/2023] Open
Abstract
Background Myeloid cells are an abundant leukocyte in many types of tumors and contribute to immune evasion. Expression of the enzyme arginase 1 (Arg1) is a defining feature of immunosuppressive myeloid cells and leads to depletion of L-arginine, a nutrient required for T cell and natural killer (NK) cell proliferation. Here we use CB-1158, a potent and orally-bioavailable small-molecule inhibitor of arginase, to investigate the role of Arg1 in regulating anti-tumor immunity. Methods CB-1158 was tested for the ability to block myeloid cell-mediated inhibition of T cell proliferation in vitro, and for tumor growth inhibition in syngeneic mouse models of cancer as a single agent and in combination with other therapies. Tumors from animals treated with CB-1158 were profiled for changes in immune cell subsets, expression of immune-related genes, and cytokines. Human tumor tissue microarrays were probed for Arg1 expression by immunohistochemistry and immunofluorescence. Cancer patient plasma samples were assessed for Arg1 protein and L-arginine by ELISA and mass spectrometry, respectively. Results CB-1158 blocked myeloid cell-mediated suppression of T cell proliferation in vitro and reduced tumor growth in multiple mouse models of cancer, as a single agent and in combination with checkpoint blockade, adoptive T cell therapy, adoptive NK cell therapy, and the chemotherapy agent gemcitabine. Profiling of the tumor microenvironment revealed that CB-1158 increased tumor-infiltrating CD8+ T cells and NK cells, inflammatory cytokines, and expression of interferon-inducible genes. Patient tumor samples from multiple histologies expressed an abundance of tumor-infiltrating Arg1+ myeloid cells. Plasma samples from cancer patients exhibited elevated Arg1 and reduced L-arginine compared to healthy volunteers. Conclusions These results demonstrate that Arg1 is a key mediator of immune suppression and that inhibiting Arg1 with CB-1158 shifts the immune landscape toward a pro-inflammatory environment, blunting myeloid cell-mediated immune evasion and reducing tumor growth. Furthermore, our results suggest that arginase blockade by CB-1158 may be an effective therapy in multiple types of cancer and combining CB-1158 with standard-of-care chemotherapy or other immunotherapies may yield improved clinical responses.
Collapse
Affiliation(s)
- Susanne M Steggerda
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA.
| | - Mark K Bennett
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Jason Chen
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Ethan Emberley
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Tony Huang
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Julie R Janes
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Weiqun Li
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Andrew L MacKinnon
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Amani Makkouk
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Gisele Marguier
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Peter J Murray
- Max Planck Institute for Biochemistry, Martinsried, Germany.,Departments of Infectious Diseases and Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Silinda Neou
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Alison Pan
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Francesco Parlati
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Mirna L M Rodriguez
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Lee-Ann Van de Velde
- Departments of Infectious Diseases and Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tracy Wang
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Melissa Works
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Jing Zhang
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Winter Zhang
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| | - Matthew I Gross
- Calithera Biosciences, 343 Oyster Point Boulevard, Suite 200, South San Francisco, CA, 94080, USA
| |
Collapse
|
45
|
Dugnani E, Pasquale V, Bordignon C, Canu A, Piemonti L, Monti P. Integrating T cell metabolism in cancer immunotherapy. Cancer Lett 2017; 411:12-18. [DOI: 10.1016/j.canlet.2017.09.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/20/2017] [Accepted: 09/24/2017] [Indexed: 01/12/2023]
|
46
|
Harris NL, Loke P. Recent Advances in Type-2-Cell-Mediated Immunity: Insights from Helminth Infection. Immunity 2017; 47:1024-1036. [DOI: 10.1016/j.immuni.2017.11.015] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 11/06/2017] [Accepted: 11/16/2017] [Indexed: 12/18/2022]
|
47
|
Lange SM, McKell MC, Schmidt SM, Hossfeld AP, Chaturvedi V, Kinder JM, McAlees JW, Lewkowich IP, Way SS, Turner J, Qualls JE. l-Citrulline Metabolism in Mice Augments CD4 + T Cell Proliferation and Cytokine Production In Vitro, and Accumulation in the Mycobacteria-Infected Lung. Front Immunol 2017; 8:1561. [PMID: 29201027 PMCID: PMC5696333 DOI: 10.3389/fimmu.2017.01561] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/31/2017] [Indexed: 12/25/2022] Open
Abstract
Activation, recruitment, and effector function of T lymphocytes are essential for control of mycobacterial infection. These processes are tightly regulated in T cells by the availability of l-arginine within the microenvironment. In turn, mycobacterial infection dampens T cell responsiveness through arginase induction in myeloid cells, promoting sequestration of l-arginine within the local milieu. Here, we show T cells can replenish intracellular l-arginine through metabolism of l-citrulline to mediate inflammatory function, allowing anti-mycobacterial T cells to overcome arginase-mediated suppression. Furthermore, T cell l-citrulline metabolism is necessary for accumulation of CD4+ T cells at the site of infection, suggesting this metabolic pathway is involved during anti-mycobacterial T cell immunity in vivo. Together, these findings establish a contribution for l-arginine synthesis by T cells during mycobacterial infection, and implicate l-citrulline as a potential immuno-nutrient to modulate host immunity.
Collapse
Affiliation(s)
- Shannon M Lange
- Laboratory of Dr. Joseph E. Qualls, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati/Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Melanie C McKell
- Laboratory of Dr. Joseph E. Qualls, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati/Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Stephanie M Schmidt
- Laboratory of Dr. Joseph E. Qualls, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Austin P Hossfeld
- Laboratory of Dr. Joanne Turner, Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Vandana Chaturvedi
- Laboratory of Dr. Sing Sing Way, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Jeremy M Kinder
- Laboratory of Dr. Sing Sing Way, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Jaclyn W McAlees
- Laboratory of Dr. Ian P. Lewkowich, Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Ian P Lewkowich
- Laboratory of Dr. Ian P. Lewkowich, Division of Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Sing Sing Way
- Laboratory of Dr. Sing Sing Way, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Joanne Turner
- Laboratory of Dr. Joanne Turner, Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States.,Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Joseph E Qualls
- Laboratory of Dr. Joseph E. Qualls, Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
48
|
Yong CS, Abba Moussa D, Cretenet G, Kinet S, Dardalhon V, Taylor N. Metabolic orchestration of T lineage differentiation and function. FEBS Lett 2017; 591:3104-3118. [PMID: 28901530 DOI: 10.1002/1873-3468.12849] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/04/2017] [Accepted: 09/06/2017] [Indexed: 12/14/2022]
Abstract
T cells are stimulated by the engagement of antigen, cytokine, pathogen, and hormone receptors. While research performed over many years has focused on deciphering the molecular components of these pathways, recent data underscore the importance of the metabolic environment in conditioning responses to receptor engagement. The ability of T cells to undergo a massive proliferation and cytokine secretion in response to receptor signals requires alterations to their bioenergetic homeostasis, allowing them to meet new energetic and biosynthetic demands. The metabolic reprogramming of activated T cells is regulated not only by changes in intracellular nutrient uptake and utilization but also by nutrient and oxygen concentrations in the extracellular environment. Notably, the extracellular environment can be profoundly altered by pathological conditions such as infections and tumors, thereby perturbing the metabolism and function of antigen-specific T lymphocytes. This review highlights the interplay between diverse metabolic networks and the transcriptional/epigenetic states that condition T-cell differentiation, comparing the metabolic features of T lymphocytes with other immune cells. We further address recent discoveries in the metabolic pathways that govern T-cell function in physiological and pathological conditions.
Collapse
Affiliation(s)
- Carmen S Yong
- IGMM, CNRS, Univ. Montpellier, Montpellier, France
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Vic., Australia
| | | | | | | | | | - Naomi Taylor
- IGMM, CNRS, Univ. Montpellier, Montpellier, France
| |
Collapse
|
49
|
Werner A, Koschke M, Leuchtner N, Luckner-Minden C, Habermeier A, Rupp J, Heinrich C, Conradi R, Closs EI, Munder M. Reconstitution of T Cell Proliferation under Arginine Limitation: Activated Human T Cells Take Up Citrulline via L-Type Amino Acid Transporter 1 and Use It to Regenerate Arginine after Induction of Argininosuccinate Synthase Expression. Front Immunol 2017; 8:864. [PMID: 28791021 PMCID: PMC5523021 DOI: 10.3389/fimmu.2017.00864] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/07/2017] [Indexed: 11/13/2022] Open
Abstract
In the tumor microenvironment, arginine is metabolized by arginase-expressing myeloid cells. This arginine depletion profoundly inhibits T cell functions and is crucially involved in tumor-induced immunosuppression. Reconstitution of adaptive immune functions in the context of arginase-mediated tumor immune escape is a promising therapeutic strategy to boost the immunological antitumor response. Arginine can be recycled in certain mammalian tissues from citrulline via argininosuccinate (ASA) in a two-step enzymatic process involving the enzymes argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL). Here, we demonstrate that anti-CD3/anti-CD28-activated human primary CD4+ and CD8+ T cells upregulate ASS expression in response to low extracellular arginine concentrations, while ASL is expressed constitutively. ASS expression peaked under moderate arginine restriction (20 µM), but no relevant induction was detectable in the complete absence of extracellular arginine. The upregulated ASS correlated with a reconstitution of T cell proliferation upon supplementation of citrulline, while the suppressed production of IFN-γ was refractory to citrulline substitution. In contrast, ASA reconstituted proliferation and cytokine synthesis even in the complete absence of arginine. By direct quantification of intracellular metabolites we show that activated primary human T cells import citrulline but only metabolize it further to ASA and arginine when ASS is expressed in the context of low amounts of extracellular arginine. We then clarify that citrulline transport is largely mediated by the L-type amino acid transporter 1 (LAT1), induced upon human T cell activation. Upon siRNA-mediated knockdown of LAT1, activated T cells lost the ability to import citrulline. These data underline the potential of citrulline substitution as a promising pharmacological way to treat immunosuppression in settings of arginine deprivation.
Collapse
Affiliation(s)
- Anke Werner
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Miriam Koschke
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nadine Leuchtner
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Luckner-Minden
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alice Habermeier
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Johanna Rupp
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christin Heinrich
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Roland Conradi
- Transfusion Center, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
50
|
Renner K, Singer K, Koehl GE, Geissler EK, Peter K, Siska PJ, Kreutz M. Metabolic Hallmarks of Tumor and Immune Cells in the Tumor Microenvironment. Front Immunol 2017; 8:248. [PMID: 28337200 PMCID: PMC5340776 DOI: 10.3389/fimmu.2017.00248] [Citation(s) in RCA: 267] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/20/2017] [Indexed: 12/14/2022] Open
Abstract
Cytotoxic T lymphocytes and NK cells play an important role in eliminating malignant tumor cells and the number and activity of tumor-infiltrating T cells represent a good marker for tumor prognosis. Based on these findings, immunotherapy, e.g., checkpoint blockade, has received considerable attention during the last couple of years. However, for the majority of patients, immune control of their tumors is gray theory as malignant cells use effective mechanisms to outsmart the immune system. Increasing evidence suggests that changes in tumor metabolism not only ensure an effective energy supply and generation of building blocks for tumor growth but also contribute to inhibition of the antitumor response. Immunosuppression in the tumor microenvironment is often based on the mutual metabolic requirements of immune cells and tumor cells. Cytotoxic T and NK cell activation leads to an increased demand for glucose and amino acids, a well-known feature shown by tumor cells. These close metabolic interdependencies result in metabolic competition, limiting the proliferation, and effector functions of tumor-specific immune cells. Moreover, not only nutrient restriction but also tumor-driven shifts in metabolite abundance and accumulation of metabolic waste products (e.g., lactate) lead to local immunosuppression, thereby facilitating tumor progression and metastasis. In this review, we describe the metabolic interplay between immune cells and tumor cells and discuss tumor cell metabolism as a target structure for cancer therapy. Metabolic (re)education of tumor cells is not only an approach to kill tumor cells directly but could overcome metabolic immunosuppression in the tumor microenvironment and thereby facilitate immunotherapy.
Collapse
Affiliation(s)
- Kathrin Renner
- Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Katrin Singer
- Internal Medicine III, University Hospital Regensburg , Regensburg , Germany
| | - Gudrun E Koehl
- Department of Surgery, University Hospital Regensburg , Regensburg , Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg , Regensburg , Germany
| | - Katrin Peter
- Internal Medicine III, University Hospital Regensburg , Regensburg , Germany
| | - Peter J Siska
- Internal Medicine III, University Hospital Regensburg , Regensburg , Germany
| | - Marina Kreutz
- Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Regensburg Center for Interventional Immunology, Regensburg, Germany
| |
Collapse
|