1
|
Ao-Di F, Han-Qing L, Xi-Zheng W, Ke Y, Hong-Xin G, Hai-Xia Z, Guan-Wei F, Li-Lan. Advances in macrophage metabolic reprogramming in myocardial ischemia-reperfusion. Cell Signal 2024; 123:111370. [PMID: 39216681 DOI: 10.1016/j.cellsig.2024.111370] [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: 07/09/2024] [Revised: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Acute myocardial infarction (AMI) is the leading cause of death worldwide, and reperfusion therapy is a critical therapeutic approach to reduce myocardial ischemic injury and minimize infarct size. However, ischemia/reperfusion (I/R) itself also causes myocardial injury, and inflammation is an essential mechanism by which it leads to myocardial injury, with macrophages as crucial immune cells in this process. Macrophages are innate immune cells that maintain tissue homeostasis, host defence during pathogen infection, and repair during tissue injury. During the acute phase of I/R, M1-type macrophages generate a pro-inflammatory milieu, clear necrotic myocardial tissue, and further recruit mononuclear (CCR2+) macrophages. Over time, the reparative (M2 type) macrophages gradually became dominant. In recent years, metabolic studies have shown a clear correlation between the metabolic profile of macrophages and their phenotype and function. M1-type macrophages are mainly characterized by glycolytic energy supply, and their tricarboxylic acid (TCA) cycle and mitochondrial oxidative phosphorylation (OXPHOS) processes are impaired. In contrast, M2 macrophages rely primarily on OXPHOS for energy. Changing the metabolic profile of macrophages can alter the macrophage phenotype. Altered energy pathways are also present in macrophages during I/R, and intervention in this process contributes to earlier and greater M2 macrophage infiltration, which may be a potential target for the treatment of myocardial I/R injury. Therefore, this paper mainly reviews the characteristics of macrophage energy metabolism alteration and phenotypic transition during I/R and its mechanism of mediating myocardial injury to provide a basis for further research in this field.
Collapse
Affiliation(s)
- Fan Ao-Di
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Han-Qing
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wang Xi-Zheng
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Ke
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guo Hong-Xin
- Heart center, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Zhang Hai-Xia
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fan Guan-Wei
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Li-Lan
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
2
|
Zwierzchowski G, Zhang G, Tobolski D, Wójcik R, Wishart DS, Ametaj BN. Metabolomic fingerprinting of milk fever cows: Pre- and postpartum metabolite alterations. J Vet Intern Med 2024. [PMID: 39466655 DOI: 10.1111/jvim.17217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 09/25/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Milk fever (MF), a metabolic disorder in dairy cows characterized by low blood calcium concentrations postpartum, is well-recognized clinically. However, comprehensive data on the alteration of metabolites associated with this condition remains sparse. HYPOTHESIS Delineate serum metabolite profiles and metabolic pathways preceding, coinciding with, and after the onset of MF. ANIMALS Twenty-six cows, including 20 healthy cows and 6 cows initially affected by MF. Because of culling, the number of MF-affected cows decreased to 4 at MF week, +4 weeks, and +8 weeks postpartum. METHODS A nested case-control longitudinal study was conducted, with blood samples collected at -8 and -4 weeks prepartum, MF week, and +4 and +8 weeks postpartum. Serum analysis utilized direct injection/liquid chromatography/tandem mass spectrometry (DI/LC/MS/MS) techniques. RESULTS Key findings included the identification of diverse metabolites such as hexose, amino acids, phosphatidylcholines, lysophosphatidylcholines, and sphingomyelin, which varied between studied groups (P < .05). The most marked metabolic alterations were observed 4 weeks prepartum. In total, 42, 56, 38, 29, and 24 metabolites distinguished the MF group at the respective time points (P < .05). Additionally, 33 metabolic pathways, including amino acid, antioxidant metabolism, fatty acid degradation, and carbohydrate processing, were impacted (P < .05). CONCLUSIONS AND CLINICAL IMPORTANCE Metabolic disruptions in dairy cows begin several weeks before the clinical manifestation of MF and persist up to 8 weeks postpartum. These findings emphasize the complexity of MF, extending beyond only hypocalcemia and indicate the necessity for preemptive monitoring in dairy herd management.
Collapse
Affiliation(s)
- Grzegorz Zwierzchowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | - Guanshi Zhang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Dawid Tobolski
- Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Roman Wójcik
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - David S Wishart
- Department of Biological and Computer Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Burim N Ametaj
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Canè S, Geiger R, Bronte V. The roles of arginases and arginine in immunity. Nat Rev Immunol 2024:10.1038/s41577-024-01098-2. [PMID: 39420221 DOI: 10.1038/s41577-024-01098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Arginase activity and arginine metabolism in immune cells have important consequences for health and disease. Their dysregulation is commonly observed in cancer, autoimmune disorders and infectious diseases. Following the initial description of a role for arginase in the dysfunction of T cells mounting an antitumour response, numerous studies have broadened our understanding of the regulation and expression of arginases and their integration with other metabolic pathways. Here, we highlight the differences in arginase compartmentalization and storage between humans and rodents that should be taken into consideration when assessing the effects of arginase activity. We detail the roles of arginases, arginine and its metabolites in immune cells and their effects in the context of cancer, autoimmunity and infectious disease. Finally, we explore potential therapeutic strategies targeting arginases and arginine.
Collapse
Affiliation(s)
- Stefania Canè
- The Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Roger Geiger
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Università della Svizzera italiana, Bellinzona, Switzerland
| | | |
Collapse
|
4
|
Aktar T, Modak S, Majumder D, Maiti D. A detailed insight into macrophages' role in shaping lung carcinogenesis. Life Sci 2024; 352:122896. [PMID: 38972632 DOI: 10.1016/j.lfs.2024.122896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
Despite significant advancements in cancer treatment in recent decades, the high mortality rate associated with lung cancer remains a significant concern. The development and proper execution of new targeted therapies needs more deep knowledge regarding the lung cancer associated tumour microenvironment. One of the key component of that tumour microenvironment is the lung resident macrophages. Although in normal physiological condition the lung resident macrophages are believed to maintain lung homeostasis, but they may also initiate a vicious inflammatory response in abnormal conditions which is linked to lung cancer development. Depending on the activation pathway, the lung resident macrophages are either of M1 or M2 sub-type. The M1 and M2 sub-types differ significantly in various prospectuses, from phenotypic markers to metabolic pathways. In addition to this generalized classification, the recent advancement of the multiomics technology is able to identify some other sub-types of lung resident macrophages. Researchers have also observed that these different sub-types can manipulate the pathogenesis of lung carcinogenesis in a context dependent manner and can either promote or inhibit the development of lung carcinogenesis upon receiving proper activation. As proper knowledge about the role played by the lung resident macrophages' in shaping the lung carcinogenesis is limited, so the main purpose of this review is to bring all the available information under the same roof. We also elaborated the different mechanisms involved in maintenance of the plasticity of M1/M2 sub-type, as this plasticity can be a good target for lung cancer treatment.
Collapse
Affiliation(s)
- Tamanna Aktar
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Snehashish Modak
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Debabrata Majumder
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India; Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Debasish Maiti
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India.
| |
Collapse
|
5
|
Xu C, Zhang J, Zhang J, Li D, Yan X, Gu Y, Zhong M, Gao H, Zhao Q, Qu X, Huang P, Zhang J. Near Infrared-Triggered Nitric Oxide-Release Nanovesicles with Mild-Photothermal Antibacterial and Immunomodulation for Healing MRSA-Infected Diabetic Wounds. Adv Healthc Mater 2024:e2402297. [PMID: 39175376 DOI: 10.1002/adhm.202402297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/05/2024] [Indexed: 08/24/2024]
Abstract
Bacterial infection-induced excessive inflammation is a major obstacle in diabetic wound healing. Nitric oxide (NO) exhibits significant antibacterial activity but is extremely deficient in diabetes. Hence, a near-infrared (NIR)-triggered NO release system is constructed through codelivery of polyarginine (PArg) and gold nanorods (Au) in an NIR-activatable methylene blue (MB) polypeptide-assembled nanovesicle (Au/PEL-PBA-MB/PArg). Upon NIR irradiation, the quenched MB in the nanovesicles is photoactivated to generate more reactive oxygen species (ROS) to oxidize PArg and release NO in an on-demand controlled manner. With the specific bacterial capture of phenylboronic acid (PBA), NO elevated membrane permeability and boosted bacterial vulnerability in the photothermal therapy (PTT) of the Au nanorods, which is displayed by superior mild PTT antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA) at temperatures < 49.7 °C in vitro. Moreover, in vivo, the antibacterial nanovesicles greatly suppressed the burst of MRSA-induced excessive inflammation, NO relayed immunomodulated macrophage polarization from M1 to M2, and the excessive inflammatory phase is successfully transferred to the repair phase. In cooperation with angiogenesis by NO, tissue regeneration is accelerated in MRSA-infected diabetic wounds. Therefore, nanoplatform has considerable potential for accelerating the healing of infected diabetic wounds.
Collapse
Affiliation(s)
- Chang Xu
- Hebei Key Laboratory of Functional Polymers, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Jiqing Zhang
- Department of Medical Ultrasound, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250000, China
| | - Junxian Zhang
- Hebei Key Laboratory of Functional Polymers, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Danting Li
- Hebei Key Laboratory of Functional Polymers, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Xiaozhe Yan
- Hebei Key Laboratory of Functional Polymers, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Yuxuan Gu
- Hebei Key Laboratory of Functional Polymers, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Meihui Zhong
- Hebei Key Laboratory of Functional Polymers, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Hui Gao
- School of Materials Science and Engineering, Tianjin University of Technology, Tianjin, 300384, China
| | - Qiang Zhao
- Key Laboratory of bioactive materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xiongwei Qu
- Hebei Key Laboratory of Functional Polymers, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Jimin Zhang
- Hebei Key Laboratory of Functional Polymers, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin, 300130, China
| |
Collapse
|
6
|
Zhang Z, Wang Y, Xia L, Zhang Y. Roles of Critical Amino Acids Metabolism in The Interactions Between Intracellular Bacterial Infection and Macrophage Function. Curr Microbiol 2024; 81:280. [PMID: 39031203 DOI: 10.1007/s00284-024-03801-x] [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: 05/03/2024] [Accepted: 07/10/2024] [Indexed: 07/22/2024]
Abstract
Macrophages, as crucial participants in the innate immune system, respond to pathogenic challenges through their dynamic metabolic adjustments, demonstrating the intimate interplay between cellular metabolism and immune function. Bacterial infection of macrophages causes changes in macrophage metabolism, affecting both macrophage function and bacterial virulence and intracellular survival. This review explores the reprogramming of amino acid metabolism in macrophages in response to bacterial infection, with a particular focus on the influence of critical amino acids such as serine, glutamine, and arginine on the immune functions of macrophages; highlights the roles of these metabolic pathways in macrophage functions such as phagocytosis, inflammatory response, immune regulation, and pathogen clearance; reveals how pathogens exploit and manipulate the amino acid metabolism within macrophages to support their own growth and replication, thereby showcasing the intricate interplay between macrophages and pathogens. It provides a foundation for understanding the interactions between macrophages amino acid metabolism and pathogens, offering potential strategies and therapeutic targets for the development of novel anti-infection therapies.
Collapse
Affiliation(s)
- Zuowei Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, 212013, Jiangsu, China
- International Genome Center, Jiangsu University, Zhenjiang, 212013, China
| | - Yurou Wang
- International Genome Center, Jiangsu University, Zhenjiang, 212013, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
7
|
Meng T, He D, Han Z, Shi R, Wang Y, Ren B, Zhang C, Mao Z, Luo G, Deng J. Nanomaterial-Based Repurposing of Macrophage Metabolism and Its Applications. NANO-MICRO LETTERS 2024; 16:246. [PMID: 39007981 PMCID: PMC11250772 DOI: 10.1007/s40820-024-01455-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024]
Abstract
Macrophage immunotherapy represents an emerging therapeutic approach aimed at modulating the immune response to alleviate disease symptoms. Nanomaterials (NMs) have been engineered to monitor macrophage metabolism, enabling the evaluation of disease progression and the replication of intricate physiological signal patterns. They achieve this either directly or by delivering regulatory signals, thereby mapping phenotype to effector functions through metabolic repurposing to customize macrophage fate for therapy. However, a comprehensive summary regarding NM-mediated macrophage visualization and coordinated metabolic rewiring to maintain phenotypic equilibrium is currently lacking. This review aims to address this gap by outlining recent advancements in NM-based metabolic immunotherapy. We initially explore the relationship between metabolism, polarization, and disease, before delving into recent NM innovations that visualize macrophage activity to elucidate disease onset and fine-tune its fate through metabolic remodeling for macrophage-centered immunotherapy. Finally, we discuss the prospects and challenges of NM-mediated metabolic immunotherapy, aiming to accelerate clinical translation. We anticipate that this review will serve as a valuable reference for researchers seeking to leverage novel metabolic intervention-matched immunomodulators in macrophages or other fields of immune engineering.
Collapse
Affiliation(s)
- Tingting Meng
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Danfeng He
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Zhuolei Han
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Rong Shi
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
- Department of Breast Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730030, People's Republic of China
| | - Yuhan Wang
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Bibo Ren
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Cheng Zhang
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China
| | - Zhengwei Mao
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China.
| | - Gaoxing Luo
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China.
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, People's Republic of China.
| |
Collapse
|
8
|
Chen C, Han P, Qing Y. Metabolic heterogeneity in tumor microenvironment - A novel landmark for immunotherapy. Autoimmun Rev 2024; 23:103579. [PMID: 39004158 DOI: 10.1016/j.autrev.2024.103579] [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: 01/31/2024] [Revised: 04/10/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The surrounding non-cancer cells and tumor cells that make up the tumor microenvironment (TME) have various metabolic rhythms. TME metabolic heterogeneity is influenced by the intricate network of metabolic control within and between cells. DNA, protein, transport, and microbial levels are important regulators of TME metabolic homeostasis. The effectiveness of immunotherapy is also closely correlated with alterations in TME metabolism. The response of a tumor patient to immunotherapy is influenced by a variety of variables, including intracellular metabolic reprogramming, metabolic interaction between cells, ecological changes within and between tumors, and general dietary preferences. Although immunotherapy and targeted therapy have made great strides, their use in the accurate identification and treatment of tumors still has several limitations. The function of TME metabolic heterogeneity in tumor immunotherapy is summarized in this article. It focuses on how metabolic heterogeneity develops and is regulated as a tumor progresses, the precise molecular mechanisms and potential clinical significance of imbalances in intracellular metabolic homeostasis and intercellular metabolic coupling and interaction, as well as the benefits and drawbacks of targeted metabolism used in conjunction with immunotherapy. This offers insightful knowledge and important implications for individualized tumor patient diagnosis and treatment plans in the future.
Collapse
Affiliation(s)
- Chen Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China
| | - Peng Han
- Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, China.
| | - Yanping Qing
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
9
|
Xu M, Cui Y, Wei S, Cong X, Chen Y, Tian S, Yao A, Chen W, Weng L. Emerging nanomaterials targeting macrophage adapted to abnormal metabolism in cancer and atherosclerosis therapy (Review). Int J Mol Med 2024; 53:13. [PMID: 38063240 PMCID: PMC10760796 DOI: 10.3892/ijmm.2023.5337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
Macrophages, as highly heterogeneous and plastic immune cells, occupy a pivotal role in both pro‑inflammatory (M1) and anti‑inflammatory (M2) responses. While M1‑type macrophages secrete pro‑inflammatory factors to initiate and sustain inflammation, M2‑type macrophages promote inflammation regression and uphold tissue homeostasis. These distinct phenotypic transitions in macrophages are closely linked to significant alterations in cellular metabolism, encompassing key response pathways such as glycolysis, pentose phosphate pathway, oxidative phosphorylation, lipid metabolism, amino acid metabolism, the tricarboxylic acid cycle and iron metabolism. These metabolic adaptations enable macrophages to adapt their activities in response to varying disease microenvironments. Therefore, the present review focused primarily on elucidating the intricate metabolic pathways that underlie macrophage functionality. Subsequently, it offers a comprehensive overview of the current state‑of‑the‑art nanomaterials, highlighting their promising potential in modulating macrophage metabolism to effectively hinder disease progression in both cancer and atherosclerosis.
Collapse
Affiliation(s)
- Miaomiao Xu
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
| | - Ying Cui
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
| | - Siyuan Wei
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
| | - Xuelong Cong
- State Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
| | - Yiying Chen
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
| | - Shujie Tian
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
| | - Anqi Yao
- RDFZ Chaoyang Branch School, Beijing 100028, P.R. China
| | - Weiwei Chen
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
| | - Lixing Weng
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
- State Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
10
|
Chen WH, Chen CH, Hsu MC, Chang RW, Wang CH, Lee TS. Advances in the molecular mechanisms of statins in regulating endothelial nitric oxide bioavailability: Interlocking biology between eNOS activity and L-arginine metabolism. Biomed Pharmacother 2024; 171:116192. [PMID: 38262153 DOI: 10.1016/j.biopha.2024.116192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
Statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A, are widely used to treat hypercholesterolemia. In addition, statins have been suggested to reduce the risk of cardiovascular events owing to their pleiotropic effects on the vascular system, including vasodilation, anti-inflammation, anti-coagulation, anti-oxidation, and inhibition of vascular smooth muscle cell proliferation. The major beneficial effect of statins in maintaining vascular homeostasis is the induction of nitric oxide (NO) bioavailability by activating endothelial NO synthase (eNOS) in endothelial cells. The mechanisms underlying the increased NO bioavailability and eNOS activation by statins have been well-established in various fields, including transcriptional and post-transcriptional regulation, kinase-dependent phosphorylation and protein-protein interactions. However, the mechanism by which statins affect the metabolism of L-arginine, a precursor of NO biosynthesis, has rarely been discussed. Autophagy, which is crucial for energy homeostasis, regulates endothelial functions, including NO production and angiogenesis, and is a potential therapeutic target for cardiovascular diseases. In this review, in addition to summarizing the molecular mechanisms underlying increased NO bioavailability and eNOS activation by statins, we also discuss the effects of statins on the metabolism of L-arginine.
Collapse
Affiliation(s)
- Wen-Hua Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Man-Chen Hsu
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ru-Wen Chang
- Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chih-Hsien Wang
- Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
11
|
Parveen S, Shen J, Lun S, Zhao L, Alt J, Koleske B, Leone RD, Rais R, Powell JD, Murphy JR, Slusher BS, Bishai WR. Glutamine metabolism inhibition has dual immunomodulatory and antibacterial activities against Mycobacterium tuberculosis. Nat Commun 2023; 14:7427. [PMID: 37973991 PMCID: PMC10654700 DOI: 10.1038/s41467-023-43304-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
As one of the most successful human pathogens, Mycobacterium tuberculosis (Mtb) has evolved a diverse array of determinants to subvert host immunity and alter host metabolic patterns. However, the mechanisms of pathogen interference with host metabolism remain poorly understood. Here we show that a glutamine metabolism antagonist, JHU083, inhibits Mtb proliferation in vitro and in vivo. JHU083-treated mice exhibit weight gain, improved survival, a 2.5 log lower lung bacillary burden at 35 days post-infection, and reduced lung pathology. JHU083 treatment also initiates earlier T-cell recruitment, increased proinflammatory myeloid cell infiltration, and a reduced frequency of immunosuppressive myeloid cells when compared to uninfected and rifampin-treated controls. Metabolomic analysis of lungs from JHU083-treated Mtb-infected mice reveals citrulline accumulation, suggesting elevated nitric oxide (NO) synthesis, and lowered levels of quinolinic acid which is derived from the immunosuppressive metabolite kynurenine. JHU083-treated macrophages also produce more NO potentiating their antibacterial activity. When tested in an immunocompromised mouse model of Mtb infection, JHU083 loses its therapeutic efficacy suggesting the drug's host-directed effects are likely to be predominant. Collectively, these data reveal that JHU083-mediated glutamine metabolism inhibition results in dual antibacterial and host-directed activity against tuberculosis.
Collapse
Affiliation(s)
- Sadiya Parveen
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jessica Shen
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shichun Lun
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Liang Zhao
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jesse Alt
- Johns Hopkins University, Baltimore, MD, USA
| | - Benjamin Koleske
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert D Leone
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan D Powell
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Calico, South San Francisco, CA, USA
| | - John R Murphy
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Barbara S Slusher
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Johns Hopkins University, Baltimore, MD, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - William R Bishai
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Xu R, Peng J, Ma Z, Xie K, Li M, Wang Q, Guo X, Nan N, Wang S, Li J, Xu J, Gong M. Prolonged administration of total glucosides of paeony improves intestinal immune imbalance and epithelial barrier damage in collagen-induced arthritis rats based on metabolomics-network pharmacology integrated analysis. Front Pharmacol 2023; 14:1187797. [PMID: 38026929 PMCID: PMC10679728 DOI: 10.3389/fphar.2023.1187797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and joint damage with complex pathological mechanisms. In recent years, many studies have shown that the dysregulation of intestinal mucosal immunity and the damage of the epithelial barrier are closely related to the occurrence of RA. Total glucosides of paeony (TGP) have been used clinically for the treatment of RA in China for decades, while the pharmacological mechanism is still uncertain. The purpose of this study was to investigate the regulatory effect and mechanism of TGP on intestinal immunity and epithelial barrier in RA model rats. The results showed that TGP alleviated immune hyperfunction by regulating the ratio of CD3+, CD4+ and CD8+ in different lymphocyte synthesis sites of the small intestine, including Peyer's patches (PPs), intraepithelial lymphocytes (IELs), and lamina propria lymphocytes (LPLs). Specially, TGP first exhibited immunomodulatory effects on sites close to the intestinal lumen (IELs and LPLs), and then on PPs far away from the intestinal lumen as the administration time prolonged. Meanwhile, TGP restores the intestinal epithelial barrier by upregulating the ratio of villi height (V)/crypt depth (C) and expression of tight junction proteins (ZO-1, occludin). Finally, the integrated analysis of metabolomics-network pharmacology was also used to explore the possible regulation mechanism of TGP on the intestinal tract. Metabolomics analysis revealed that TGP reversed the intestinal metabolic profile disturbance in CIA rats, and identified 32 biomarkers and 163 corresponding targets; network pharmacology analysis identified 111 potential targets for TGP to treat RA. By intersecting the results of the two, three key targets such as ADA, PNP and TYR were determined. Pharmacological verification experiments showed that the levels of ADA and PNP in the small intestine of CIA rats were significantly increased, while TGP significantly decreased their ADA and PNP levels. In conclusion, purine metabolism may play an important role in the process of TGP improving RA-induced intestinal immune imbalance and impaired epithelial barrier.
Collapse
Affiliation(s)
- Rui Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Jine Peng
- Department of Pharmacy, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhe Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Kaili Xie
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Meijing Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Qi Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Xiaomeng Guo
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Nan Nan
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Sihui Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Jing Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Jingjing Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| | - Muxin Gong
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, China
| |
Collapse
|
13
|
Hissen KL, He W, Wu G, Criscitiello MF. Immunonutrition: facilitating mucosal immune response in teleost intestine with amino acids through oxidant-antioxidant balance. Front Immunol 2023; 14:1241615. [PMID: 37841275 PMCID: PMC10570457 DOI: 10.3389/fimmu.2023.1241615] [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: 06/19/2023] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
Comparative animal models generate fundamental scientific knowledge of immune responses. However, these studies typically are conducted in mammals because of their biochemical and physiological similarity to humans. Presently, there has been an interest in using teleost fish models to study intestinal immunology, particularly intestinal mucosa immune response. Instead of targeting the pathogen itself, a preferred approach for managing fish health is through nutrient supplementation, as it is noninvasive and less labor intensive than vaccine administrations while still modulating immune properties. Amino acids (AAs) regulate metabolic processes, oxidant-antioxidant balance, and physiological requirements to improve immune response. Thus, nutritionists can develop sustainable aquafeeds through AA supplementation to promote specific immune responses, including the intestinal mucosa immune system. We propose the use of dietary supplementation with functional AAs to improve immune response by discussing teleost fish immunology within the intestine and explore how oxidative burst is used as an immune defense mechanism. We evaluate immune components and immune responses in the intestine that use oxidant-antioxidant balance through potential selection of AAs and their metabolites to improve mucosal immune capacity and gut integrity. AAs are effective modulators of teleost gut immunity through oxidant-antioxidant balance. To incorporate nutrition as an immunoregulatory means in teleost, we must obtain more tools including genomic, proteomic, nutrition, immunology, and macrobiotic and metabonomic analyses, so that future studies can provide a more holistic understanding of the mucosal immune system in fish.
Collapse
Affiliation(s)
- Karina L. Hissen
- Comparative Immunogenetics Laboratory Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Wenliang He
- Amino Acid Laboratory, Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Guoyao Wu
- Amino Acid Laboratory, Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Michael F. Criscitiello
- Comparative Immunogenetics Laboratory Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M Health Science Center, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
14
|
Ruocco C, Malavazos AE, Ragni M, Carruba MO, Valerio A, Iacobellis G, Nisoli E. Amino acids contribute to adaptive thermogenesis. New insights into the mechanisms of action of recent drugs for metabolic disorders are emerging. Pharmacol Res 2023; 195:106892. [PMID: 37619907 DOI: 10.1016/j.phrs.2023.106892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/28/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Adaptive thermogenesis is the heat production by muscle contractions (shivering thermogenesis) or brown adipose tissue (BAT) and beige fat (non-shivering thermogenesis) in response to external stimuli, including cold exposure. BAT and beige fat communicate with peripheral organs and the brain through a variegate secretory and absorption processes - controlling adipokines, microRNAs, extracellular vesicles, and metabolites - and have received much attention as potential therapeutic targets for managing obesity-related disorders. The sympathetic nervous system and norepinephrine-releasing adipose tissue macrophages (ATM) activate uncoupling protein 1 (UCP1), expressed explicitly in brown and beige adipocytes, dissolving the electrochemical gradient and uncoupling tricarboxylic acid cycle and the electron transport chain from ATP production. Mounting evidence has attracted attention to the multiple effects of dietary and endogenously synthesised amino acids in BAT thermogenesis and metabolic phenotype in animals and humans. However, the mechanisms implicated in these processes have yet to be conclusively characterized. In the present review article, we aim to define the principal investigation areas in this context, including intestinal microbiota constitution, adipose autophagy modulation, and secretome and metabolic fluxes control, which lead to increased brown/beige thermogenesis. Finally, also based on our recent epicardial adipose tissue results, we summarise the evidence supporting the notion that the new dual and triple agonists of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon (GCG) receptor - with never before seen weight loss and insulin-sensitizing efficacy - promote thermogenic-like amino acid profiles in BAT with robust heat production and likely trigger sympathetic activation and adaptive thermogenesis by controlling amino acid metabolism and ATM expansion in BAT and beige fat.
Collapse
Affiliation(s)
- Chiara Ruocco
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Alexis Elias Malavazos
- Endocrinology Unit, Clinical Nutrition and Cardiovascular Prevention Service, IRCCS Policlinico San Donato, Piazza Edmondo Malan, 2, San Donato Milanese, 20097 Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, via della Commenda, 10, 20122 Milan, Italy
| | - Maurizio Ragni
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Michele O Carruba
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa, 11, 25123 Brescia, Italy
| | - Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami, 1400 NW 12th Ave, Miami, FL, USA
| | - Enzo Nisoli
- Center for Study and Research on Obesity, Department of Biomedical Technology and Translational Medicine, University of Milan, via Vanvitelli, 32, 20129 Milan, Italy.
| |
Collapse
|
15
|
Ou Y, Yang Y, Li X, Zhang X, Zhao L, Yang C, Wu Y. Arginine metabolism key enzymes affect the prognosis of myelodysplastic syndrome by interfering with macrophage polarization. Cancer Med 2023; 12:16444-16454. [PMID: 37366304 PMCID: PMC10469818 DOI: 10.1002/cam4.6287] [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: 02/18/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
INTRODUCTION Immune factors contribute to the onset of myelodysplastic syndrome (MDS). Arginine metabolism affects tumor-associated macrophage (TAM) polarization. This study investigated the infiltration of TAMs and effect of arginine metabolism key enzymes on MDS prognosis. METHODS We used the GEO (Gene Express Omnibus database) dataset "GSE19429" to analyze and compare metabolism-associated pathways between MDS patients with excess blasts and those without. The markers of TAMs and arginine metabolism key enzymes, including CD68, iNOS, ARG1 and ASS1 were included in this study. A cohort of 79 patients with acute myeloid leukemia or MDS extracted from GenomicScape's online data mining platform was used to analyze the prognostic significance of the mRNA levels. Fifty-eight patients with primary MDS admitted to Sichuan University's West China Hospital from 2013 to 2017 were evaluated for protein levels. The coexpression of CD68, iNOS, and ARG1 was investigated using an Opal polychromatic immunofluorescence kit. RESULTS The "Arginine and proline metabolism" pathways (padjusted = 0.01) were associated with excess blasts in patients with MDS. In the mRNA expression cohort, patients with low NOS2 (or iNOS) and high ARG1, ASS1, and CD68 expression levels had worse prognosis. Patients with high CD68 (p = 0.01), high iNOS (p < 0.01), low ARG1 (p = 0.01), and negative ASS1 (p = 0.02) protein expression levels had better prognoses. iNOS and ARG1 were coexpressed with CD68 in MDS patients with or without excess blasts, respectively. CONCLUSIONS Arginine metabolism may contribute to the prognosis of patients with MDS by affecting TAM polarization.
Collapse
Affiliation(s)
- Yang Ou
- Department of Hematology and Hematology Research InstituteWest China Hospital, Sichuan UniversityChengduPeople's Republic of China
| | - Yan Yang
- Department of Hematology and Hematology Research InstituteWest China Hospital, Sichuan UniversityChengduPeople's Republic of China
| | - Xuefeng Li
- Department of Hematology and Hematology Research InstituteWest China Hospital, Sichuan UniversityChengduPeople's Republic of China
| | - Xin Zhang
- Department of Hematology and Hematology Research InstituteWest China Hospital, Sichuan UniversityChengduPeople's Republic of China
| | - Lei Zhao
- Department of Hematology and Hematology Research InstituteWest China Hospital, Sichuan UniversityChengduPeople's Republic of China
| | - Chenlu Yang
- Department of Hematology and Hematology Research InstituteWest China Hospital, Sichuan UniversityChengduPeople's Republic of China
| | - Yu Wu
- Department of Hematology and Hematology Research InstituteWest China Hospital, Sichuan UniversityChengduPeople's Republic of China
| |
Collapse
|
16
|
Chen S, Saeed AFUH, Liu Q, Jiang Q, Xu H, Xiao GG, Rao L, Duo Y. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther 2023; 8:207. [PMID: 37211559 DOI: 10.1038/s41392-023-01452-1] [Citation(s) in RCA: 312] [Impact Index Per Article: 312.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 05/23/2023] Open
Abstract
Macrophages exist in various tissues, several body cavities, and around mucosal surfaces and are a vital part of the innate immune system for host defense against many pathogens and cancers. Macrophages possess binary M1/M2 macrophage polarization settings, which perform a central role in an array of immune tasks via intrinsic signal cascades and, therefore, must be precisely regulated. Many crucial questions about macrophage signaling and immune modulation are yet to be uncovered. In addition, the clinical importance of tumor-associated macrophages is becoming more widely recognized as significant progress has been made in understanding their biology. Moreover, they are an integral part of the tumor microenvironment, playing a part in the regulation of a wide variety of processes including angiogenesis, extracellular matrix transformation, cancer cell proliferation, metastasis, immunosuppression, and resistance to chemotherapeutic and checkpoint blockade immunotherapies. Herein, we discuss immune regulation in macrophage polarization and signaling, mechanical stresses and modulation, metabolic signaling pathways, mitochondrial and transcriptional, and epigenetic regulation. Furthermore, we have broadly extended the understanding of macrophages in extracellular traps and the essential roles of autophagy and aging in regulating macrophage functions. Moreover, we discussed recent advances in macrophages-mediated immune regulation of autoimmune diseases and tumorigenesis. Lastly, we discussed targeted macrophage therapy to portray prospective targets for therapeutic strategies in health and diseases.
Collapse
Affiliation(s)
- Shanze Chen
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Abdullah F U H Saeed
- Department of Cancer Biology, Beckman Research Institute of City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Quan Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen University, Shenzhen, 518052, China
| | - Qiong Jiang
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Haizhao Xu
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- Department of Respiratory, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Gary Guishan Xiao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian, China.
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Yanhong Duo
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
17
|
Starikova EA, Rubinstein AA, Mammedova JT, Isakov DV, Kudryavtsev IV. Regulated Arginine Metabolism in Immunopathogenesis of a Wide Range of Diseases: Is There a Way to Pass between Scylla and Charybdis? Curr Issues Mol Biol 2023; 45:3525-3551. [PMID: 37185755 PMCID: PMC10137093 DOI: 10.3390/cimb45040231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
More than a century has passed since arginine was discovered, but the metabolism of the amino acid never ceases to amaze researchers. Being a conditionally essential amino acid, arginine performs many important homeostatic functions in the body; it is involved in the regulation of the cardiovascular system and regeneration processes. In recent years, more and more facts have been accumulating that demonstrate a close relationship between arginine metabolic pathways and immune responses. This opens new opportunities for the development of original ways to treat diseases associated with suppressed or increased activity of the immune system. In this review, we analyze the literature describing the role of arginine metabolism in the immunopathogenesis of a wide range of diseases, and discuss arginine-dependent processes as a possible target for therapeutic approaches.
Collapse
Affiliation(s)
- Eleonora A Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L'va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Artem A Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Jennet T Mammedova
- Laboratory of General Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Dmitry V Isakov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L'va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Igor V Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- School of Biomedicine, Far Eastern Federal University, FEFU Campus, 10 Ajax Bay, Russky Island, 690922 Vladivostok, Russia
| |
Collapse
|
18
|
Parveen S, Shen J, Lun S, Zhao L, Koleske B, Leone RD, Rais R, Powell JD, Murphy JR, Slusher BS, Bishai WR. Glutamine metabolism inhibition has dual immunomodulatory and antibacterial activities against Mycobacterium tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529704. [PMID: 36865287 PMCID: PMC9980128 DOI: 10.1101/2023.02.23.529704] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
As one of the most successful human pathogens, Mycobacterium tuberculosis (Mtb) has evolved a diverse array of determinants to subvert host immunity and alter host metabolic patterns. However, the mechanisms of pathogen interference with host metabolism remain poorly understood. Here we show that a novel glutamine metabolism antagonist, JHU083, inhibits Mtb proliferation in vitro and in vivo. JHU083-treated mice exhibit weight gain, improved survival, a 2.5 log lower lung bacillary burden at 35 days post-infection, and reduced lung pathology. JHU083 treatment also initiates earlier T-cell recruitment, increased proinflammatory myeloid cell infiltration, and a reduced frequency of immunosuppressive myeloid cells when compared to uninfected and rifampin-treated controls. Metabolomics analysis of lungs from JHU083-treated Mtb-infected mice revealed reduced glutamine levels, citrulline accumulation suggesting elevated NOS activity, and lowered levels of quinolinic acid which is derived from the immunosuppressive metabolite kynurenine. When tested in an immunocompromised mouse model of Mtb infection, JHU083 lost its therapeutic efficacy suggesting the drug's host-directed effects are likely to be predominant. Collectively, these data reveal that JHU083-mediated glutamine metabolism inhibition results in dual antibacterial and host-directed activity against tuberculosis.
Collapse
Affiliation(s)
- Sadiya Parveen
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Jessica Shen
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Shichun Lun
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Liang Zhao
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Benjamin Koleske
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Robert D. Leone
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jonathan D. Powell
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - John R. Murphy
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - William R. Bishai
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
19
|
Bouabid C, Rabhi S, Thedinga K, Barel G, Tnani H, Rabhi I, Benkahla A, Herwig R, Guizani-Tabbane L. Host M-CSF induced gene expression drives changes in susceptible and resistant mice-derived BMdMs upon Leishmania major infection. Front Immunol 2023; 14:1111072. [PMID: 37187743 PMCID: PMC10175952 DOI: 10.3389/fimmu.2023.1111072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Leishmaniases are a group of diseases with different clinical manifestations. Macrophage-Leishmania interactions are central to the course of the infection. The outcome of the disease depends not only on the pathogenicity and virulence of the parasite, but also on the activation state, the genetic background, and the underlying complex interaction networks operative in the host macrophages. Mouse models, with mice strains having contrasting behavior in response to parasite infection, have been very helpful in exploring the mechanisms underlying differences in disease progression. We here analyzed previously generated dynamic transcriptome data obtained from Leishmania major (L. major) infected bone marrow derived macrophages (BMdMs) from resistant and susceptible mouse. We first identified differentially expressed genes (DEGs) between the M-CSF differentiated macrophages derived from the two hosts, and found a differential basal transcriptome profile independent of Leishmania infection. These host signatures, in which 75% of the genes are directly or indirectly related to the immune system, may account for the differences in the immune response to infection between the two strains. To gain further insights into the underlying biological processes induced by L. major infection driven by the M-CSF DEGs, we mapped the time-resolved expression profiles onto a large protein-protein interaction (PPI) network and performed network propagation to identify modules of interacting proteins that agglomerate infection response signals for each strain. This analysis revealed profound differences in the resulting responses networks related to immune signaling and metabolism that were validated by qRT-PCR time series experiments leading to plausible and provable hypotheses for the differences in disease pathophysiology. In summary, we demonstrate that the host's gene expression background determines to a large degree its response to L. major infection, and that the gene expression analysis combined with network propagation is an effective approach to help identifying dynamically altered mouse strain-specific networks that hold mechanistic information about these contrasting responses to infection.
Collapse
Affiliation(s)
- Cyrine Bouabid
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
- Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Sameh Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Kristina Thedinga
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Gal Barel
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Hedia Tnani
- Laboratory de BioInformatic, BioMathematic and BioStatistic (BIMS), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Imen Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
- Higher Institute of Biotechnology at Sidi-Thabet (ISBST), Biotechnopole Sidi-Thabet- University of Manouba, Sidi-Thabet, Tunisia
| | - Alia Benkahla
- Laboratory de BioInformatic, BioMathematic and BioStatistic (BIMS), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Ralf Herwig
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Lamia Guizani-Tabbane
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
- *Correspondence: Lamia Guizani-Tabbane,
| |
Collapse
|
20
|
DIK I, HATIPOGLU D, GULERSOY E. Comparison of some cytokines, acute phase proteins and citrulline levels in healthy and canine distemper infected dogs. J Vet Med Sci 2023; 85:76-82. [PMID: 36418074 PMCID: PMC9887225 DOI: 10.1292/jvms.22-0281] [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] [Indexed: 11/23/2022] Open
Abstract
Canine distemper virus (CDV) is the etiological agent of severe disease in domestic and wild carnivores. Clinical diagnosis of CDV is challenging because of its similarity to other canine respiratory and intestinal diseases. We aimed to determine certain cytokine (interleukin [IL]-1β, IL-2, IL-4, IL-6, IL-10, and tumor necrosis factor-α [TNF-α]), interferon (IFN)-γ, canine serum amyloid A (SAA), and canine citrulline (CIT) levels for the first time in CDV-positive dogs. For this purpose, 10 CDV-positive dogs with compatible clinical findings (i.e., neurological symptoms such as tremors and myoclonus, ocular and nasal discharge, and wheezing) and 10 healthy dogs based on the clinical examinations and rapid test results were enrolled. It was observed that the CIT, INF-γ, IL-1β, IL-2, IL-6, and TNF-α levels were significantly decreased in the CDV-positive dogs than that of the healthy ones (P<0.05). As a result, it was observed that CDV causes immunosuppression and accordingly, the inflammatory response might cause decreased cytokine and acute-phase protein synthesis. Therefore, it was concluded that further investigation of inflammatory pathways and CIT interactions may provide crucial clinical information at different stages of CDV, and aforementioned parameters may serve as important biomarkers for CDV in terms of demonstrating the presence of immunosuppression.
Collapse
Affiliation(s)
- Irmak DIK
- Department of Virology, Faculty of Veterinary Medicine, University of Selcuk, Konya, Türkiye
| | - Durmus HATIPOGLU
- Department of Physiology, Faculty of Veterinary Medicine, University of Selcuk, Konya, Türkiye,Correspondence to: Hatıpoglu D: , Department of Virology, Faculty of Veterinary Medicine, University of Selcuk, 42075, Konya, Türkiye
| | - Erdem GULERSOY
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Harran, Şanlıurfa, Türkiye
| |
Collapse
|
21
|
Novoselova AV, Yushina MN, Patysheva MR, Prostashkina EA, Bragina OD, Garbukov EY, Kzhyshkowska JG. Peculiarities of amino acid profile in monocytes in breast cancer. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2022. [DOI: 10.24075/brsmu.2022.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Monocytes are large circulating white blood cells that are the main precursors of tissue macrophages as well as tumor-associated macrophages in the adult body. Different types of monocytes have multidirectional effects on the growth and metastatic spread of cancer cells, both activating and inhibiting these processes. Tumor progression is associated with the triggering of a whole cascade of inflammatory and immune reactions. These pathological processes are associated with changes in the amino acid content of monocytes, which can lead to disruption of their function, in particular their migration, division and maturation. The aim of the work was to profile the amino acids of monocytes, followed by a study of the amino acid composition of monocytes from patients with breast cancer using liquid chromatography with mass spectrometric detection. Significant differences in metabolite levels in monocytes of breast cancer patients and monocytes of healthy donors were found for glycine (p-value = 0.0127), asparagine (p-value = 0.0197), proline (p-value = 0.0159), methionine (p-value = 0.0357), tryptophan (p-value = 0.0028), tyrosine (p-value = 0.0127). In the study, we identified biological networks that could potentially be involved in altering the phenotype of monocytes affected by breast cancer (BC), using bioinformatic analysis of metabolic pathways involving the discovered amino acids. Mathematical models based on amino acid combinations with 100% sensitivity and specificity have been developed. Features of immune system cell metabolism in BC have been identified and potential diagnostic biomarkers have been proposed.
Collapse
Affiliation(s)
- AV Novoselova
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - MN Yushina
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - MR Patysheva
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk, Russia; Tomsk National State University, Tomsk, Russia
| | - EA Prostashkina
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - OD Bragina
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk, Russia
| | - EYu Garbukov
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk, Russia
| | - JG Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
22
|
Park HE, Lee W, Choi S, Jung M, Shin MK, Shin SJ. Modulating macrophage function to reinforce host innate resistance against Mycobacterium avium complex infection. Front Immunol 2022; 13:931876. [PMID: 36505429 PMCID: PMC9730288 DOI: 10.3389/fimmu.2022.931876] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 10/21/2022] [Indexed: 11/25/2022] Open
Abstract
Mycobacterium avium complex (MAC) is the main causative agent of infectious diseases in humans among nontuberculous mycobacteria (NTM) that are ubiquitous organisms found in environmental media such as soil as well as in domestic and natural waters. MAC is a primary causative agent of NTM-lung disease that threaten immunocompromised or structural lung disease patients. The incidence and the prevalence of M. tuberculosis infection have been reduced, while MAC infections and mortality rates have increased, making it a cause of global health concern. The emergence of drug resistance and the side effects of long-term drug use have led to a poor outcome of treatment regimens against MAC infections. Therefore, the development of host-directed therapy (HDT) has recently gained interest, aiming to accelerate mycobacterial clearance and reversing lung damage by employing the immune system using a novel adjuvant strategy to improve the clinical outcome of MAC infection. Therefore, in this review, we discuss the innate immune responses that contribute to MAC infection focusing on macrophages, chief innate immune cells, and host susceptibility factors in patients. We also discuss potential HDTs that can act on the signaling pathway of macrophages, thereby contributing to antimycobacterial activity as a part of the innate immune response during MAC infection. Furthermore, this review provides new insights into MAC infection control that modulates and enhances macrophage function, promoting host antimicrobial activity in response to potential HDTs and thus presenting a deeper understanding of the interactions between macrophages and MACs during infection.
Collapse
Affiliation(s)
- Hyun-Eui Park
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sangwon Choi
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Myunghwan Jung
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea,*Correspondence: Min-Kyoung Shin, ; Sung Jae Shin,
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea,*Correspondence: Min-Kyoung Shin, ; Sung Jae Shin,
| |
Collapse
|
23
|
Ashokcoomar S, Reedoy KS, Loots DT, Beukes D, van Reenen M, Pillay B, Pillay M. M. tuberculosis curli pili (MTP) facilitates a reduction of microbicidal activity of infected THP-1 macrophages during early stages of infection. Comp Immunol Microbiol Infect Dis 2022; 90-91:101907. [DOI: 10.1016/j.cimid.2022.101907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
|
24
|
Snyder BM, Gebretsadik T, Turi KN, McKennan C, Havstad S, Jackson DJ, Ober C, Lynch S, McCauley K, Seroogy CM, Zoratti EM, Khurana Hershey GK, Berdnikovs S, Cunningham G, Summar ML, Gern JE, Hartert TV. Association of citrulline concentration at birth with lower respiratory tract infection in infancy: Findings from a multi-site birth cohort study. Front Pediatr 2022; 10:979777. [PMID: 36324820 PMCID: PMC9618869 DOI: 10.3389/fped.2022.979777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
Assessing the association of the newborn metabolic state with severity of subsequent respiratory tract infection may provide important insights on infection pathogenesis. In this multi-site birth cohort study, we identified newborn metabolites associated with lower respiratory tract infection (LRTI) in the first year of life in a discovery cohort and assessed for replication in two independent cohorts. Increased citrulline concentration was associated with decreased odds of LRTI (discovery cohort: aOR 0.83 [95% CI 0.70-0.99], p = 0.04; replication cohorts: aOR 0.58 [95% CI 0.28-1.22], p = 0.15). While our findings require further replication and investigation of mechanisms of action, they identify a novel target for LRTI prevention and treatment.
Collapse
Affiliation(s)
- Brittney M. Snyder
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tebeb Gebretsadik
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kedir N. Turi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christopher McKennan
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Suzanne Havstad
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, United States
| | - Daniel J. Jackson
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, United States
| | - Susan Lynch
- Department of Medicine, University of California, San Francisco, CA, United States
| | - Kathryn McCauley
- Department of Medicine, University of California, San Francisco, CA, United States
| | | | - Edward M. Zoratti
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States
| | - Gurjit K. Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Sergejs Berdnikovs
- Department of Medicine, Northwestern University, Chicago, IL, United States
| | - Gary Cunningham
- Department of Genetics and Metabolism, Children’s National Medical Center, Washington, DC, United States
| | - Marshall L. Summar
- Department of Genetics and Metabolism, Children’s National Medical Center, Washington, DC, United States
| | - James E. Gern
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
| | - Tina V. Hartert
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | | |
Collapse
|
25
|
Wei Z, Oh J, Flavell RA, Crawford JM. LACC1 bridges NOS2 and polyamine metabolism in inflammatory macrophages. Nature 2022; 609:348-353. [PMID: 35978195 PMCID: PMC9813773 DOI: 10.1038/s41586-022-05111-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 07/14/2022] [Indexed: 01/11/2023]
Abstract
The mammalian immune system uses various pattern recognition receptors to recognize invaders and host damage and transmits this information to downstream immunometabolic signalling outcomes. Laccase domain-containing 1 (LACC1) protein is an enzyme highly expressed in inflammatory macrophages and serves a central regulatory role in multiple inflammatory diseases such as inflammatory bowel diseases, arthritis and clearance of microbial infection1-4. However, the biochemical roles required for LACC1 functions remain largely undefined. Here we elucidated a shared biochemical function of LACC1 in mice and humans, converting L-citrulline to L-ornithine (L-Orn) and isocyanic acid and serving as a bridge between proinflammatory nitric oxide synthase (NOS2) and polyamine immunometabolism. We validated the genetic and mechanistic connections among NOS2, LACC1 and ornithine decarboxylase 1 (ODC1) in mouse models and bone marrow-derived macrophages infected by Salmonella enterica Typhimurium. Strikingly, LACC1 phenotypes required upstream NOS2 and downstream ODC1, and Lacc1-/- chemical complementation with its product L-Orn significantly restored wild-type activities. Our findings illuminate a previously unidentified pathway in inflammatory macrophages, explain why its deficiency may contribute to human inflammatory diseases and suggest that L-Orn could serve as a nutraceutical to ameliorate LACC1-associated immunological dysfunctions such as arthritis or inflammatory bowel disease.
Collapse
Affiliation(s)
- Zheng Wei
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, CT, USA
| | - Joonseok Oh
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, CT, USA
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA.
| | - Jason M Crawford
- Institute of Biomolecular Design & Discovery, Yale University, West Haven, CT, USA.
- Department of Chemistry, Yale University, New Haven, CT, USA.
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
Sagawa CHD, Assis RDAB, Zaini PA, Saxe H, Wilmarth PA, Salemi M, Phinney BS, Dandekar AM. De Novo Arginine Synthesis Is Required for Full Virulence of Xanthomonas arboricola pv. juglandis During Walnut Bacterial Blight Disease. PHYTOPATHOLOGY 2022; 112:1500-1512. [PMID: 34941365 DOI: 10.1094/phyto-07-21-0302-r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Walnut blight (WB) disease caused by Xanthomonas arboricola pv. juglandis (Xaj) threatens orchards worldwide. Nitrogen metabolism in this bacterial pathogen is dependent on arginine, a nitrogen-enriched amino acid that can either be synthesized or provided by the plant host. The arginine biosynthetic pathway uses argininosuccinate synthase (argG), associated with increased bacterial virulence. We examined the effects of bacterial arginine and nitrogen metabolism on the plant response during WB by proteomic analysis of the mutant strain Xaj argG-. Phenotypically, the mutant strain produced 42% fewer symptoms and survived in the plant tissue with 2.5-fold reduced growth compared with wild type, while showing itself to be auxotrophic for arginine in vitro. Proteomic analysis of infected tissue enabled the profiling of 676 Xaj proteins and 3,296 walnut proteins using isobaric labeling in a data-dependent acquisition approach. Comparative analysis of differentially expressed proteins revealed distinct plant responses. Xaj wild type (WT) triggered processes of catabolism and oxidative stress in the host under observed disease symptoms, while most of the host biosynthetic processes triggered by Xaj WT were inhibited during Xaj argG- infection. Overall, the Xaj proteins revealed a drastic shift in carbon and energy management induced by disruption of nitrogen metabolism while the top differentially expressed proteins included a Fis transcriptional regulator and a peptidyl-prolyl isomerase. Our results show the critical role of de novo arginine biosynthesis to sustain virulence and minimal growth during WB. This study is timely and critical as copper-based control methods are losing their effectiveness, and new sustainable methods are urgently needed in orchard environments.[Formula: see text] Copyright © 2022 The Author(s). This is an open access article distributed under the CC BY-NC-ND 4.0 International license.
Collapse
Affiliation(s)
- Cíntia H D Sagawa
- Department of Plant Sciences, University of California, Davis, CA 95616, U.S.A
| | - Renata de A B Assis
- Department of Plant Sciences, University of California, Davis, CA 95616, U.S.A
- Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG 35400-000, Brazil
| | - Paulo A Zaini
- Department of Plant Sciences, University of California, Davis, CA 95616, U.S.A
| | - Houston Saxe
- Department of Plant Sciences, University of California, Davis, CA 95616, U.S.A
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, U.S.A
| | - Michelle Salemi
- Proteomics Core Facility, University of California, Davis, CA 95616, U.S.A
| | - Brett S Phinney
- Proteomics Core Facility, University of California, Davis, CA 95616, U.S.A
| | - Abhaya M Dandekar
- Department of Plant Sciences, University of California, Davis, CA 95616, U.S.A
| |
Collapse
|
27
|
Glutamine Is Required for M1-like Polarization of Macrophages in Response to Mycobacterium tuberculosis Infection. mBio 2022; 13:e0127422. [PMID: 35762591 PMCID: PMC9426538 DOI: 10.1128/mbio.01274-22] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In response to Mycobacterium tuberculosis infection, macrophages mount proinflammatory and antimicrobial responses similar to those observed in M1 macrophages activated by lipopolysaccharide (LPS) and interferon gamma (IFN-γ). A metabolic reprogramming to hypoxia-inducible-factor 1 (HIF-1)-mediated uptake of glucose and its metabolism by glycolysis is required for M1-like polarization, but little is known about other metabolic programs driving the M1-like polarization during infection. We report that glutamine serves as a carbon and nitrogen source for the metabolic reprogramming to M1-like macrophages. Widely targeted metabolite screening identified an association of glutamine and/or glutamate with highly affected metabolic pathways of M1-like macrophages. Moreover, stable isotope-assisted metabolomics of U13C glutamine and U13C glucose revealed that glutamine, rather than glucose, is catabolized in both the oxidative and reductive tricarboxylic acid (TCA) cycles of M1-like macrophages, thereby generating signaling molecules that include succinate, biosynthetic precursors such as aspartate, and itaconate. U15N glutamine-tracing metabolomics further revealed participation of glutamine nitrogen in synthesis of intermediates of purine and pyrimidine metabolism plus amino acids, including aspartate. These findings were corroborated by diminished M1 polarization from chemical inhibition of glutaminase (GLS), the key enzyme in the glutaminolysis pathway, and by genetic deletion of GLS in infected macrophages. Thus, the catabolism of glutamine is an integral component of metabolic reprogramming in activating macrophages and it coordinates with elevated cytosolic glycolysis to satisfy the cellular demand for bioenergetic and biosynthetic precursors of M1-like macrophages. Knowledge of these new immunometabolic features of M1-like macrophages should advance the development of host-directed therapies for tuberculosis.
Collapse
|
28
|
Micallef P, Vujičić M, Wu Y, Peris E, Wang Y, Chanclón B, Ståhlberg A, Cardell SL, Wernstedt Asterholm I. C1QTNF3 is Upregulated During Subcutaneous Adipose Tissue Remodeling and Stimulates Macrophage Chemotaxis and M1-Like Polarization. Front Immunol 2022; 13:914956. [PMID: 35720277 PMCID: PMC9202579 DOI: 10.3389/fimmu.2022.914956] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/06/2022] [Indexed: 01/08/2023] Open
Abstract
The adipose tissue undergoes substantial tissue remodeling during weight gain-induced expansion as well as in response to the mechanical and immunological stresses from a growing tumor. We identified the C1q/TNF-related protein family member C1qtnf3 as one of the most upregulated genes that encode secreted proteins in tumor-associated inguinal adipose tissue - especially in high fat diet-induced obese mice that displayed 3-fold larger tumors than their lean controls. Interestingly, inguinal adipose tissue C1qtnf3 was co-regulated with several macrophage markers and chemokines and was primarily expressed in fibroblasts while only low levels were detected in adipocytes and macrophages. Administration of C1QTNF3 neutralizing antibodies inhibited macrophage accumulation in tumor-associated inguinal adipose tissue while tumor growth was unaffected. In line with this finding, C1QTNF3 exerted chemotactic actions on both M1- and M2-polarized macrophages in vitro. Moreover, C1QTNF3 treatment of M2-type macrophages stimulated the ERK and Akt pathway associated with increased M1-like polarization as judged by increased expression of M1-macrophage markers, increased production of nitric oxide, reduced oxygen consumption and increased glycolysis. Based on these results, we propose that macrophages are recruited to adipose tissue sites with increased C1QTNF3 production. However, the impact of the immunomodulatory effects of C1QTNF3 in adipose tissue remodeling warrants future investigations.
Collapse
Affiliation(s)
- Peter Micallef
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Milica Vujičić
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Yanling Wu
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Eduard Peris
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Ying Wang
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Belén Chanclón
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Göteborg, Sweden.,Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Susanna L Cardell
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
29
|
Magdalena D, Michal S, Marta S, Magdalena KP, Anna P, Magdalena G, Rafał S. Targeted metabolomics analysis of serum and Mycobacterium tuberculosis antigen-stimulated blood cultures of pediatric patients with active and latent tuberculosis. Sci Rep 2022; 12:4131. [PMID: 35260782 PMCID: PMC8904507 DOI: 10.1038/s41598-022-08201-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/03/2022] [Indexed: 12/28/2022] Open
Abstract
Profound tuberculosis (TB)-induced metabolic changes reflected in the blood metabolomic profile may provide an opportunity to identify specific markers of Mycobacterium tuberculosis infection. Using targeted liquid chromatography tandem mass spectrometry, we compared the levels of 30 small metabolites, including amino acids and derivatives, and small organic compounds in serum and M.tb antigen-stimulated whole blood cultures of active TB children, latent TB (LTBI) children, nonmycobacterial pneumonia (NMP) children, and healthy controls (HCs) to assess their potential as biomarkers of childhood TB. We found elevated levels of leucine and kynurenine combined with reduced concentrations of citrulline and glutamine in serum and blood cultures of TB and LTBI groups. LTBI status was additionally associated with a decrease in valine levels in blood cultures. The NMP metabolite profile was characterized by an increase in citrulline and glutamine and a decrease in leucine, kynurenine and valine concentrations. The highest discriminatory potential for identifying M.tb infection was observed for leucine detected in serum and kynurenine in stimulated blood cultures. The use of targeted metabolomics may reveal metabolic changes in M.tb-infected children, and the obtained results are a proof of principle of the usefulness of metabolites in the auxiliary diagnosis of TB in children.
Collapse
Affiliation(s)
- Druszczynska Magdalena
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland.
| | - Seweryn Michal
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland
| | | | - Kowalewska-Pietrzak Magdalena
- Regional Specialized Hospital of Tuberculosis, Lung Diseases, and Rehabilitation in Lodz, Okolna 181, 91-520, Lodz, Poland
| | - Pankowska Anna
- Regional Specialized Hospital of Tuberculosis, Lung Diseases, and Rehabilitation in Lodz, Okolna 181, 91-520, Lodz, Poland
| | - Godkowicz Magdalena
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland
| | - Szewczyk Rafał
- , Labexperts sp z o. o. Piekarnicza 5, 80-126, Gdansk, Poland
| |
Collapse
|
30
|
Gauthier T, Chen W. Modulation of Macrophage Immunometabolism: A New Approach to Fight Infections. Front Immunol 2022; 13:780839. [PMID: 35154105 PMCID: PMC8825490 DOI: 10.3389/fimmu.2022.780839] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Macrophages are essential innate immune cells that contribute to host defense during infection. An important feature of macrophages is their ability to respond to extracellular cues and to adopt different phenotypes and functions in response to these stimuli. The evidence accumulated in the last decade has highlighted the crucial role of metabolic reprogramming during macrophage activation in infectious context. Thus, understanding and manipulation of macrophage immunometabolism during infection could be of interest to develop therapeutic strategies. In this review, we focus on 5 major metabolic pathways including glycolysis, pentose phosphate pathway, fatty acid oxidation and synthesis, tricarboxylic acid cycle and amino acid metabolism and discuss how they sustain and regulate macrophage immune function in response to parasitic, bacterial and viral infections as well as trained immunity. At the end, we assess whether some drugs including those used in clinic and in development can target macrophage immunometabolism for potential therapy during infection with an emphasis on SARS-CoV2 infection.
Collapse
Affiliation(s)
- Thierry Gauthier
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Wanjun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
31
|
Crowther RR, Schmidt SM, Lange SM, McKell MC, Robillard MC, Zhao J, Haffey WD, Wyder MA, Greis KD, Setchell KDR, Qualls JE. Cutting Edge: l-Arginine Transfer from Antigen-Presenting Cells Sustains CD4 + T Cell Viability and Proliferation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:793-798. [PMID: 35101895 PMCID: PMC8820592 DOI: 10.4049/jimmunol.2100652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/01/2021] [Indexed: 11/19/2022]
Abstract
Metabolomics analyses suggest changes in amino acid abundance, particularly l-arginine (L-ARG), occur in patients with tuberculosis. Immune cells require L-ARG to fuel effector functions following infection. We have previously described an L-ARG synthesis pathway in immune cells; however, its role in APCs has yet to be uncovered. Using a coculture system with mycobacterial-specific CD4+ T cells, we show APC L-ARG synthesis supported T cell viability and proliferation, and activated T cells contained APC-derived L-ARG. We hypothesize that APCs supply L-ARG to support T cell activation under nutrient-limiting conditions. This work expands the current model of APC-T cell interactions and provides insight into the effects of nutrient availability in immune cells.
Collapse
Affiliation(s)
- Rebecca R Crowther
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Stephanie M Schmidt
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Shannon M Lange
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Melanie C McKell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Michelle C Robillard
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Junfang Zhao
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Wendy D Haffey
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH; and
- Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Michael A Wyder
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH; and
- Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kenneth D Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH; and
- Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kenneth D R Setchell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Joseph E Qualls
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH;
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
32
|
Mao Y, Shi D, Li G, Jiang P. Citrulline depletion by ASS1 is required for proinflammatory macrophage activation and immune responses. Mol Cell 2021; 82:527-541.e7. [PMID: 35016033 DOI: 10.1016/j.molcel.2021.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/22/2021] [Accepted: 12/08/2021] [Indexed: 12/21/2022]
Abstract
Citrulline can be converted into argininosuccinate by argininosuccinate synthetase (ASS1) in the urea cycle and the citrulline-nitric oxide cycle. However, the regulation and biological function of citrulline metabolism remain obscure in the immune system. Unexpectedly, we found that macrophage citrulline declines rapidly after interferon gamma (IFN-γ) and/or lipopolysaccharide (LPS) stimulation, which is required for efficient proinflammatory signaling activation. Mechanistically, IFN-γ and/or LPS stimulation promotes signal transducers and activators of transcription 1 (STAT1)-mediated ASS1 transcription and Janus kinase2 (JAK2)-mediated phosphorylation of ASS1 at tyrosine 87, thereby leading to citrulline depletion. Reciprocally, increased citrulline directly binds to JAK2 and inhibits JAK2-STAT1 signaling. Blockage of ASS1-mediated citrulline depletion suppresses the host defense against bacterial infection in vivo. We therefore define a central role for ASS1 in controlling inflammatory macrophage activation and antibacterial defense through depletion of cellular citrulline and, further, identify citrulline as an innate immune-signaling metabolite that engages a metabolic checkpoint for proinflammatory responses.
Collapse
Affiliation(s)
- Youxiang Mao
- Tsinghua-Peking Center for Life Sciences, 100084 Beijing, China; School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Di Shi
- Tsinghua-Peking Center for Life Sciences, 100084 Beijing, China; School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Gen Li
- Tsinghua-Peking Center for Life Sciences, 100084 Beijing, China; School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Peng Jiang
- Tsinghua-Peking Center for Life Sciences, 100084 Beijing, China; School of Life Sciences, Tsinghua University, 100084 Beijing, China.
| |
Collapse
|
33
|
Umar S, Palasiewicz K, Volin MV, Zanotti B, Al-Awqati M, Sweiss N, Shahrara S. IRAK4 inhibitor mitigates joint inflammation by rebalancing metabolism malfunction in RA macrophages and fibroblasts. Life Sci 2021; 287:120114. [PMID: 34732329 PMCID: PMC10020992 DOI: 10.1016/j.lfs.2021.120114] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 11/28/2022]
Abstract
Recent studies show a connection between glycolysis and inflammatory response in rheumatoid arthritis (RA) macrophages (MΦs) and fibroblasts (FLS). Yet, it is unclear which pathways could be targeted to rebalance RA MΦs and FLS metabolic reprogramming. To identify novel targets that could normalize RA metabolic reprogramming, TLR7-mediated immunometabolism was characterized in RA MΦs, FLS and experimental arthritis. We uncovered that GLUT1, HIF1α, cMYC, LDHA and lactate were responsible for the TLR7-potentiated metabolic rewiring in RA MΦs and FLS, which was negated by IRAK4i. While in RA FLS, HK2 was uniquely expanded by TLR7 and negated by IRAK4i. Conversely, TLR7-driven hypermetabolism, non-oxidative PPP (CARKL) and oxidative phosphorylation (PPARγ) were narrowly dysregulated in TLR7-activated RA MΦs and FLS and was reversed by IRAK4i. Consistently, IRAK4i therapy disrupted arthritis mediated by miR-Let7b/TLR7 along with impairing a broad-range of glycolytic intermediates, GLUT1, HIF1α, cMYC, HK2, PFKFB3, PKM2, PDK1 and RAPTOR. Notably, inhibition of the mutually upregulated glycolytic metabolites, HIF1α and cMYC, was capable of mitigating TLR7-induced inflammatory imprint in RA MΦs and FLS. In keeping with IRAK4i, treatment with HIF1i and cMYCi intercepted TLR7-enhanced IRF5 and IRF7 in RA MΦs, distinct from RA FLS. Interestingly, in RA MΦs and FLS, IRAK4i counteracted TLR7-induced CARKL reduction in line with HIF1i. Whereas, cMYCi in concordance with IRAK4i, overturned oxidative phosphorylation via PPARγ in TLR7-activated RA MΦs and FLS. The blockade of IRAK4 and its interconnected intermediates can rebalance the metabolic malfunction by obstructing glycolytic and inflammatory phenotypes in RA MΦs and FLS.
Collapse
Affiliation(s)
- Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States of America; Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, IL 60612, United States of America
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States of America; Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, IL 60612, United States of America
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, United States of America
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, United States of America
| | - Mina Al-Awqati
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States of America; Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, IL 60612, United States of America
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, IL 60612, United States of America
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL 60612, United States of America; Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, IL 60612, United States of America.
| |
Collapse
|
34
|
Mhandire K, Saggu K, Buxbaum NP. Immunometabolic Therapeutic Targets of Graft-versus-Host Disease (GvHD). Metabolites 2021; 11:736. [PMID: 34822394 PMCID: PMC8619522 DOI: 10.3390/metabo11110736] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 01/17/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative option in the treatment of aggressive malignant and non-malignant blood disorders. However, the benefits of allo-HSCT can be compromised by graft-versus-host disease (GvHD), a prevalent and morbid complication of allo-HSCT. GvHD occurs when donor immune cells mount an alloreactive response against host antigens due to histocompatibility differences between the donor and host, which may result in extensive tissue injury. The reprogramming of cellular metabolism is a feature of GvHD that is associated with the differentiation of donor CD4+ cells into the pathogenic Th1 and Th17 subsets along with the dysfunction of the immune-suppressive protective T regulatory cells (Tregs). The activation of glycolysis and glutaminolysis with concomitant changes in fatty acid oxidation metabolism fuel the anabolic activities of the proliferative alloreactive microenvironment characteristic of GvHD. Thus, metabolic therapies such as glycolytic enzyme inhibitors and fatty acid metabolism modulators are a promising therapeutic strategy for GvHD. We comprehensively review the role of cellular metabolism in GvHD pathogenesis, identify candidate therapeutic targets, and describe potential strategies for augmenting immunometabolism to ameliorate GvHD.
Collapse
|
35
|
Chen R, Chen L. Solute carrier transporters: emerging central players in tumour immunotherapy. Trends Cell Biol 2021; 32:186-201. [PMID: 34511324 DOI: 10.1016/j.tcb.2021.08.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023]
Abstract
Solute carrier transporters (SLCs) mediate nutrient and metabolite cellular homeostasis. Immune cells depend on SLCs to induce rapid and robust metabolic reprogramming, thereby controlling diverse immunological responses. Recent studies hint toward an important role of SLCs in immunity. Here, we review the emerging roles of SLCs in immunotherapy via modifying the metabolism and effector functions of immune cells. We focus on the roles of three major nutrient (glucose, amino acid, and lipid)-related transporters in immunity of representative cells [T cells, dendritic cells (DCs), natural killer (NK) cells, and macrophages) in innate and adaptive immunity. Other SLCs, such as ion transporters are also briefly discussed. Finally, we propose some potential strategies for targeting SLCs to augment tumour immunotherapy.
Collapse
Affiliation(s)
- Ruiqun Chen
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Ligong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China; Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
36
|
Luan H, Horng T. Dynamic changes in macrophage metabolism modulate induction and suppression of Type I inflammatory responses. Curr Opin Immunol 2021; 73:9-15. [PMID: 34399114 DOI: 10.1016/j.coi.2021.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 07/13/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022]
Abstract
During microbial infection, macrophages link recognition of microbial stimuli to the induction of Type I inflammatory responses. Such inflammatory responses coordinate host defense and pathogen elimination but induce significant tissue damage if sustained, so macrophages are initially activated to induce inflammatory responses but then shift to a tolerant state to suppress inflammatory responses. Macrophage tolerance is regulated by induction of negative regulators of TLR signaling, but its metabolic basis was not known. Here, we review recent studies that indicate that macrophage metabolism changes dynamically over the course of microbial exposure to influence a shift in the inflammatory response. In particular, an initial increase in oxidative metabolism boosts the induction of inflammatory responses, but is followed by a shutdown of oxidative metabolism that contributes to suppression of inflammatory responses. We propose a unifying model for how dynamic changes to oxidative metabolism influences regulation of macrophage inflammatory responses during microbial exposure.
Collapse
Affiliation(s)
- Haoming Luan
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| | - Tiffany Horng
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
37
|
Appiah MG, Park EJ, Akama Y, Nakamori Y, Kawamoto E, Gaowa A, Shimaoka M. Cellular and Exosomal Regulations of Sepsis-Induced Metabolic Alterations. Int J Mol Sci 2021; 22:ijms22158295. [PMID: 34361061 PMCID: PMC8347112 DOI: 10.3390/ijms22158295] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a sustained systemic inflammatory condition involving multiple organ failures caused by dysregulated immune response to infections. Sepsis induces substantial changes in energy demands at the cellular level leading to metabolic reprogramming in immune cells and stromal cells. Although sepsis-associated organ dysfunction and mortality have been partly attributed to the initial acute hyperinflammation and immunosuppression precipitated by a dysfunction in innate and adaptive immune responses, the late mortality due to metabolic dysfunction and immune paralysis currently represent the major problem in clinics. It is becoming increasingly recognized that intertissue and/or intercellular metabolic crosstalk via endocrine factors modulates maintenance of homeostasis, and pathological events in sepsis and other inflammatory diseases. Exosomes have emerged as a novel means of intercellular communication in the regulation of cellular metabolism, owing to their capacity to transfer bioactive payloads such as proteins, lipids, and nucleic acids to their target cells. Recent evidence demonstrates transfer of intact metabolic intermediates from cancer-associated fibroblasts via exosomes to modify metabolic signaling in recipient cells and promote cancer progression. Here, we review the metabolic regulation of endothelial cells and immune cells in sepsis and highlight the role of exosomes as mediators of cellular metabolic signaling in sepsis.
Collapse
Affiliation(s)
- Michael G. Appiah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan; (M.G.A.); (Y.A.); (Y.N.); (E.K.); (A.G.)
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan; (M.G.A.); (Y.A.); (Y.N.); (E.K.); (A.G.)
- Correspondence: (E.J.P.); (M.S.); Tel.: +81-59-231-6408 (E.J.P.); +81-59-231-5036 (M.S.)
| | - Yuichi Akama
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan; (M.G.A.); (Y.A.); (Y.N.); (E.K.); (A.G.)
- Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan
| | - Yuki Nakamori
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan; (M.G.A.); (Y.A.); (Y.N.); (E.K.); (A.G.)
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan; (M.G.A.); (Y.A.); (Y.N.); (E.K.); (A.G.)
- Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan
| | - Arong Gaowa
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan; (M.G.A.); (Y.A.); (Y.N.); (E.K.); (A.G.)
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City 514-8507, Mie, Japan; (M.G.A.); (Y.A.); (Y.N.); (E.K.); (A.G.)
- Correspondence: (E.J.P.); (M.S.); Tel.: +81-59-231-6408 (E.J.P.); +81-59-231-5036 (M.S.)
| |
Collapse
|
38
|
Rasheed A, Rayner KJ. Macrophage Responses to Environmental Stimuli During Homeostasis and Disease. Endocr Rev 2021; 42:407-435. [PMID: 33523133 PMCID: PMC8284619 DOI: 10.1210/endrev/bnab004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/20/2022]
Abstract
Work over the last 40 years has described macrophages as a heterogeneous population that serve as the frontline surveyors of tissue immunity. As a class, macrophages are found in almost every tissue in the body and as distinct populations within discrete microenvironments in any given tissue. During homeostasis, macrophages protect these tissues by clearing invading foreign bodies and/or mounting immune responses. In addition to varying identities regulated by transcriptional programs shaped by their respective environments, macrophage metabolism serves as an additional regulator to temper responses to extracellular stimuli. The area of research known as "immunometabolism" has been established within the last decade, owing to an increase in studies focusing on the crosstalk between altered metabolism and the regulation of cellular immune processes. From this research, macrophages have emerged as a prime focus of immunometabolic studies, although macrophage metabolism and their immune responses have been studied for centuries. During disease, the metabolic profile of the tissue and/or systemic regulators, such as endocrine factors, become increasingly dysregulated. Owing to these changes, macrophage responses can become skewed to promote further pathophysiologic changes. For instance, during diabetes, obesity, and atherosclerosis, macrophages favor a proinflammatory phenotype; whereas in the tumor microenvironment, macrophages elicit an anti-inflammatory response to enhance tumor growth. Herein we have described how macrophages respond to extracellular cues including inflammatory stimuli, nutrient availability, and endocrine factors that occur during and further promote disease progression.
Collapse
Affiliation(s)
- Adil Rasheed
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Katey J Rayner
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
39
|
Jiang Q, Shi L. Coordination of the Uptake and Metabolism of Amino Acids in Mycobacterium tuberculosis-Infected Macrophages. Front Immunol 2021; 12:711462. [PMID: 34326848 PMCID: PMC8315098 DOI: 10.3389/fimmu.2021.711462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/25/2021] [Indexed: 01/31/2023] Open
Abstract
Macrophage polarization to the M1-like phenotype, which is critical for the pro-inflammatory and antimicrobial responses of macrophages against intracellular pathogens, is associated with metabolic reprogramming to the Warburg effect and a high output of NO from increased expression of NOS2. However, there is limited understanding about the uptake and metabolism of other amino acids during M1 polarization. Based on functional analysis of a group of upregulated transporters and enzymes involved in the uptake and/or metabolism of amino acids in Mycobacterium tuberculosis-infected macrophages, plus studies of immune cell activation, we postulate a coherent scheme for amino acid uptake and metabolism during macrophage polarization to the M1-like phenotype. We describe potential mechanisms that the increased arginine metabolism by NOS2 is metabolically coupled with system L transporters LAT1 and LAT2 for the uptake of neutral amino acids, including those that drive mTORC1 signaling toward the M1-like phenotype. We also discuss the underappreciated pleiotropic roles of glutamine metabolism in the metabolic reprogramming of M1-like macrophages. Collectively, our analyses argue that a coordinated amino acid uptake and metabolism constitutes an integral component of the broad metabolic scheme required for macrophage polarization to M1-like phenotype against M. tuberculosis infection. This idea could stimulate future experimental efforts to elucidate the metabolic map of macrophage activation for the development of anti-tuberculosis therapies.
Collapse
Affiliation(s)
- Qingkui Jiang
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers The State University of New Jersey, Newark, NJ, United States
| | - Lanbo Shi
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
40
|
Martí I Líndez AA, Reith W. Arginine-dependent immune responses. Cell Mol Life Sci 2021; 78:5303-5324. [PMID: 34037806 PMCID: PMC8257534 DOI: 10.1007/s00018-021-03828-4] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
A growing body of evidence indicates that, over the course of evolution of the immune system, arginine has been selected as a node for the regulation of immune responses. An appropriate supply of arginine has long been associated with the improvement of immune responses. In addition to being a building block for protein synthesis, arginine serves as a substrate for distinct metabolic pathways that profoundly affect immune cell biology; especially macrophage, dendritic cell and T cell immunobiology. Arginine availability, synthesis, and catabolism are highly interrelated aspects of immune responses and their fine-tuning can dictate divergent pro-inflammatory or anti-inflammatory immune outcomes. Here, we review the organismal pathways of arginine metabolism in humans and rodents, as essential modulators of the availability of this semi-essential amino acid for immune cells. We subsequently review well-established and novel findings on the functional impact of arginine biosynthetic and catabolic pathways on the main immune cell lineages. Finally, as arginine has emerged as a molecule impacting on a plethora of immune functions, we integrate key notions on how the disruption or perversion of arginine metabolism is implicated in pathologies ranging from infectious diseases to autoimmunity and cancer.
Collapse
Affiliation(s)
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
41
|
McKell MC, Crowther RR, Schmidt SM, Robillard MC, Cantrell R, Lehn MA, Janssen EM, Qualls JE. Promotion of Anti-Tuberculosis Macrophage Activity by L-Arginine in the Absence of Nitric Oxide. Front Immunol 2021; 12:653571. [PMID: 34054815 PMCID: PMC8160513 DOI: 10.3389/fimmu.2021.653571] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/26/2021] [Indexed: 12/31/2022] Open
Abstract
Macrophages are indispensable immune cells tasked at eliminating intracellular pathogens. Mycobacterium tuberculosis (Mtb), one of the most virulent intracellular bacterial pathogens known to man, infects and resides within macrophages. While macrophages can be provoked by extracellular stimuli to inhibit and kill Mtb bacilli, these host defense mechanisms can be blocked by limiting nutritional metabolites, such as amino acids. The amino acid L-arginine has been well described to enhance immune function, especially in the context of driving macrophage nitric oxide (NO) production in mice. In this study, we aimed to establish the necessity of L-arginine on anti-Mtb macrophage function independent of NO. Utilizing an in vitro system, we identified that macrophages relied on NO for only half of their L-arginine-mediated host defenses and this L-arginine-mediated defense in the absence of NO was associated with enhanced macrophage numbers and viability. Additionally, we observed macrophage glycolysis to be driven by both L-arginine and mechanistic target of rapamycin (mTOR), and inhibition of glycolysis or mTOR reduced macrophage control of Mtb as well as macrophage number and viability in the presence of L-arginine. Our data underscore L-arginine as an essential nutrient for macrophage function, not only by fueling anti-mycobacterial NO production, but also as a central regulator of macrophage metabolism and additional host defense mechanisms.
Collapse
Affiliation(s)
- Melanie C McKell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Rebecca R Crowther
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Stephanie M Schmidt
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Michelle C Robillard
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Rachel Cantrell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Maria A Lehn
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Edith M Janssen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Joseph E Qualls
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
42
|
Abstract
Macrophages are instrumental for the repair of organs that become injured due to ischemia, yet their potential for healing is sensitive to the availability of metabolites from the surrounding milieu. This sensitivity extends beyond anabolic and catabolic reactions, as metabolites are also leveraged to control production of secreted factors that direct intercellular crosstalk. In response to limiting extracellular oxygen, acute-phase macrophages activate hypoxia-inducible transcription factors that repurpose cellular metabolism. Subsequent repair-phase macrophages secrete cytokines to activate stromal cells, the latter which contribute to matrix deposition and scarring. As we now appreciate, these distinct functions are calibrated by directing flux of carbons and cofactors into specific metabolic shunts. This occurs through glycolysis, the pentose phosphate shunt, the tricarboxylic acid cycle, oxidative phosphorylation, nicotinamide adenine dinucleotides, lipids, amino acids, and through lesser understood pathways. The integration of metabolism with macrophage function is particularly important during injury to the ischemic heart, as glucose and lipid imbalance lead to inefficient repair and permanent loss of non-regenerative muscle. Here we review macrophage metabolic signaling under ischemic stress with implications for cardiac repair.
Collapse
Affiliation(s)
- Edward B Thorp
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
43
|
Roy DG, Kaymak I, Williams KS, Ma EH, Jones RG. Immunometabolism in the Tumor Microenvironment. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2021. [DOI: 10.1146/annurev-cancerbio-030518-055817] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Advances in immunotherapy have underscored the importance of antitumor immune responses in controlling cancer. However, the tumor microenvironment (TME) imposes several obstacles to the proper function of immune cells, including a metabolically challenging and immunosuppressive microenvironment. The increased metabolic activity of tumor cells can lead to the depletion of key nutrients required by immune cells and the accumulation of byproducts that hamper antitumor immunity. Furthermore, the presence of suppressive immune cells, such as regulatory T cells and myeloid-derived suppressor cells, and the expression of immune inhibitory receptors can negatively impact immune cell metabolism and function. This review summarizes the metabolic reprogramming that is characteristic of various immune cell subsets, discusses how the metabolism and function of immune cells are shaped by the TME, and highlights how therapeutic interventions aimed at improving the metabolic fitness of immune cells and alleviating the metabolic constraints in the TME can boost antitumor immunity.
Collapse
Affiliation(s)
- Dominic G. Roy
- Goodman Cancer Research Centre, Faculty of Medicine, McGill University, Montreal, Quebec H3A 1A3, Canada
| | - Irem Kaymak
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| | - Kelsey S. Williams
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| | - Eric H. Ma
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| | - Russell G. Jones
- Metabolic and Nutritional Programming, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| |
Collapse
|
44
|
Complexity of macrophage metabolism in infection. Curr Opin Biotechnol 2021; 68:231-239. [PMID: 33610128 DOI: 10.1016/j.copbio.2021.01.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 01/14/2021] [Accepted: 01/28/2021] [Indexed: 12/23/2022]
Abstract
Macrophages are the prominent innate immune cells to combat infection and then restore tissue homeostasis after clearance of pathogens. Intracellular metabolic reprogramming is required for macrophage activation and function, as such adaptations confer macrophages with sufficient energy and metabolites to support biosynthesis and diverse functions. During the last 10 years, knowledge in this field has been greatly extended by outstanding advances demonstrating that several metabolic intermediates possess the ability to directly control macrophage activation and effector functions by various mechanisms. Of note, citrate and succinate contribute to the inflammatory activation of macrophages while tricarboxylic acid cycle-derived metabolite itaconate has a variety of immunomodulatory effects. Such progress not only encourages a further exploration into the emerging new area immunometabolism, but also provides potential therapeutic targets to control unwanted inflammation due to infection.
Collapse
|
45
|
Crowther RR, Qualls JE. Metabolic Regulation of Immune Responses to Mycobacterium tuberculosis: A Spotlight on L-Arginine and L-Tryptophan Metabolism. Front Immunol 2021; 11:628432. [PMID: 33633745 PMCID: PMC7900187 DOI: 10.3389/fimmu.2020.628432] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/30/2020] [Indexed: 12/16/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a leading cause of death worldwide. Despite decades of research, there is still much to be uncovered regarding the immune response to Mtb infection. Here, we summarize the current knowledge on anti-Mtb immunity, with a spotlight on immune cell amino acid metabolism. Specifically, we discuss L-arginine and L-tryptophan, focusing on their requirements, regulatory roles, and potential use as adjunctive therapy in TB patients. By continuing to uncover the immune cell contribution during Mtb infection and how amino acid utilization regulates their functions, it is anticipated that novel host-directed therapies may be developed and/or refined, helping to eradicate TB.
Collapse
Affiliation(s)
- Rebecca R Crowther
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Joseph E Qualls
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
46
|
Bertero E, Dudek J, Cochain C, Delgobo M, Ramos G, Gerull B, Higuchi T, Vaeth M, Zernecke A, Frantz S, Hofmann U, Maack C. Immuno-metabolic interfaces in cardiac disease and failure. Cardiovasc Res 2021; 118:37-52. [PMID: 33537710 DOI: 10.1093/cvr/cvab036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/01/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
The interplay between the cardiovascular system, metabolism, and inflammation plays a central role in the pathophysiology of a wide spectrum of cardiovascular diseases, including heart failure. Here, we provide an overview of the fundamental aspects of the interrelation between inflammation and metabolism, ranging from the role of metabolism in immune cell function to the processes how inflammation modulates systemic and cardiac metabolism. Furthermore, we discuss how disruption of this immuno-metabolic interface is involved in the development and progression of cardiovascular disease, with a special focus on heart failure. Finally, we present new technologies and therapeutic approaches that have recently emerged and hold promise for the future of cardiovascular medicine.
Collapse
Affiliation(s)
- Edoardo Bertero
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Hospital Würzburg, Germany
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Hospital Würzburg, Germany
| | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany.,Comprehensive Heart Failure Center (CHFC), Würzburg, Germany
| | - Murilo Delgobo
- Comprehensive Heart Failure Center (CHFC), Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Germany
| | - Gustavo Ramos
- Comprehensive Heart Failure Center (CHFC), Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Germany
| | - Brenda Gerull
- Department of Internal Medicine I, University Hospital Würzburg, Germany.,Department of Cardiovascular Genetics, CHFC, University Hospital Würzburg, Germany
| | - Takahiro Higuchi
- Comprehensive Heart Failure Center (CHFC), Würzburg, Germany.,Department of Nuclear Medicine, University Hospital Würzburg, Germany
| | - Martin Vaeth
- Institute of Systems Immunology, Julius-Maximilians University Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany
| | - Stefan Frantz
- Comprehensive Heart Failure Center (CHFC), Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Germany
| | - Ulrich Hofmann
- Comprehensive Heart Failure Center (CHFC), Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Hospital Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Germany
| |
Collapse
|
47
|
Liu Y, Xu R, Gu H, Zhang E, Qu J, Cao W, Huang X, Yan H, He J, Cai Z. Metabolic reprogramming in macrophage responses. Biomark Res 2021; 9:1. [PMID: 33407885 PMCID: PMC7786975 DOI: 10.1186/s40364-020-00251-y] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022] Open
Abstract
Macrophages are critical mediators of tissue homeostasis, with the function of tissue development and repair, but also in defense against pathogens. Tumor-associated macrophages (TAMs) are considered as the main component in the tumor microenvironment and play an important role in tumor initiation, growth, invasion, and metastasis. Recently, metabolic studies have revealeded specific metabolic pathways in macrophages are tightly associated with their phenotype and function. Generally, pro-inflammatory macrophages (M1) rely mainly on glycolysis and exhibit impairment of the tricarboxylic acid (TCA) cycle and mitochondrial oxidative phosphorylation (OXPHOS), whereas anti-inflammatory macrophages (M2) are more dependent on mitochondrial OXPHOS. However, accumulating evidence suggests that macrophage metabolism is not as simple as previously thought. This review discusses recent advances in immunometabolism and describes how metabolism determines macrophage phenotype and function. In addition, we describe the metabolic characteristics of TAMs as well as their therapeutic implications. Finally, we discuss recent obstacles facing this area as well as promising directions for future study.
Collapse
Affiliation(s)
- Yang Liu
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Ruyi Xu
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Huiyao Gu
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Jianwei Qu
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Wen Cao
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Xi Huang
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Haimeng Yan
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Afliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China. .,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China. .,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
| |
Collapse
|
48
|
Gobert AP, Finley JL, Latour YL, Asim M, Smith TM, Verriere TG, Barry DP, Allaman MM, Delagado AG, Rose KL, Calcutt MW, Schey KL, Sierra JC, Piazuelo MB, Mirmira RG, Wilson KT. Hypusination Orchestrates the Antimicrobial Response of Macrophages. Cell Rep 2020; 33:108510. [PMID: 33326776 PMCID: PMC7812972 DOI: 10.1016/j.celrep.2020.108510] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 10/28/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
Innate responses of myeloid cells defend against pathogenic bacteria via inducible effectors. Deoxyhypusine synthase (DHPS) catalyzes the transfer of the N-moiety of spermidine to the lysine-50 residue of eukaryotic translation initiation factor 5A (EIF5A) to form the amino acid hypusine. Hypusinated EIF5A (EIF5AHyp) transports specific mRNAs to ribosomes for translation. We show that DHPS is induced in macrophages by two gastrointestinal pathogens, Helicobacter pylori and Citrobacter rodentium, resulting in enhanced hypusination of EIF5A. EIF5AHyp was also increased in gastric macrophages from patients with H. pylori gastritis. Furthermore, we identify the bacteria-induced immune effectors regulated by hypusination. This set of proteins includes essential constituents of antimicrobial response and autophagy. Mice with myeloid cell-specific deletion of Dhps exhibit reduced EIF5AHyp in macrophages and increased bacterial burden and inflammation. Thus, regulation of translation through hypusination is a critical hallmark of the defense of eukaryotic hosts against pathogenic bacteria.
Collapse
Affiliation(s)
- Alain P Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Jordan L Finley
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yvonne L Latour
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Thaddeus M Smith
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Thomas G Verriere
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Daniel P Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Margaret M Allaman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alberto G Delagado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kristie L Rose
- Department of Biochemistry, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - M Wade Calcutt
- Department of Biochemistry, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kevin L Schey
- Department of Biochemistry, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Johanna C Sierra
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - M Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Raghavendra G Mirmira
- Translational Research Center, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37232, USA.
| |
Collapse
|
49
|
Bok R, Guerra DD, Lorca RA, Wennersten SA, Harris PS, Rauniyar AK, Stabler SP, MacLean KN, Roede JR, Brown LD, Hurt KJ. Cystathionine γ-lyase promotes estrogen-stimulated uterine artery blood flow via glutathione homeostasis. Redox Biol 2020; 40:101827. [PMID: 33485059 PMCID: PMC7823052 DOI: 10.1016/j.redox.2020.101827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/16/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022] Open
Abstract
During pregnancy, estrogen (E2) stimulates uterine artery blood flow (UBF) by enhancing nitric oxide (NO)-dependent vasodilation. Cystathionine γ-lyase (CSE) promotes vascular NO signaling by producing hydrogen sulfide (H2S) and by maintaining the ratio of reduced-to-oxidized intracellular glutathione (GSH/GSSG) through l-cysteine production. Because redox homeostasis can influence NO signaling, we hypothesized that CSE mediates E2 stimulation of UBF by modulating local intracellular cysteine metabolism and GSH/GSSG levels to promote redox homeostasis. Using non-pregnant ovariectomized WT and CSE-null (CSE KO) mice, we performed micro-ultrasound of mouse uterine and renal arteries to assess changes in blood flow upon exogenous E2 stimulation. We quantified serum and uterine artery NO metabolites (NOx), serum amino acids, and uterine and renal artery GSH/GSSG. WT and CSE KO mice exhibited similar baseline uterine and renal blood flow. Unlike WT, CSE KO mice did not exhibit expected E2 stimulation of UBF. Renal blood flow was E2-insensitive for both genotypes. While serum and uterine artery NOx were similar between genotypes at baseline, E2 decreased NOx in CSE KO serum. Cysteine was also lower in CSE KO serum, while citrulline and homocysteine levels were elevated. E2 and CSE deletion additively decreased GSH/GSSG in uterine arteries. In contrast, renal artery GSH/GSSG was insensitive to E2 or CSE deletion. Together, these findings suggest that CSE maintenance of uterine artery GSH/GSSG facilitates nitrergic signaling in uterine arteries and is required for normal E2 stimulation of UBF. These data have implications for pregnancy pathophysiology and the selective hormone responses of specific vascular beds. CSE-null mice exhibit abnormal estrogen augmentation of uterine artery blood flow. Estrogen lowers uterine artery nitric oxide metabolites in CSE null mice. CSE loss and estrogen additively impair uterine artery glutathione homeostasis. Neither CSE loss nor estrogen influences renal artery blood flow or glutathione.
Collapse
Affiliation(s)
- Rachael Bok
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Aurora, CO, 80045, USA
| | - Damian D Guerra
- Department of Biology, University of Louisville, 2301 S. 3rd Street, Louisville, KY, 40292, USA
| | - Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Aurora, CO, 80045, USA
| | - Sara A Wennersten
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Aurora, CO, 80045, USA
| | - Peter S Harris
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Blvd, Aurora, CO, 80045, USA
| | - Abhishek K Rauniyar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Blvd, Aurora, CO, 80045, USA
| | - Sally P Stabler
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Aurora, CO, 80045, USA
| | - Kenneth N MacLean
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Aurora, CO, 80045, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Blvd, Aurora, CO, 80045, USA
| | - Laura D Brown
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Perinatal Research Center, 13243 E. 23rd Avenue, Aurora, CO, 80045, USA
| | - K Joseph Hurt
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Aurora, CO, 80045, USA; Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
50
|
Daibert RMDP, de Biagi Junior CAO, Vieira FDO, da Silva MVGB, Hottz ED, Mendonça Pinheiro MB, Faza DRDLR, Pereira HP, Martins MF, Brandão HDM, Machado MA, Carvalho WA. Lipopolysaccharide triggers different transcriptional signatures in taurine and indicine cattle macrophages: Reactive oxygen species and potential outcomes to the development of immune response to infections. PLoS One 2020; 15:e0241861. [PMID: 33156842 PMCID: PMC7647108 DOI: 10.1371/journal.pone.0241861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Macrophages are classified upon activation as classical activated M1 and M2 anti-inflammatory regulatory populations. This macrophage polarization is well characterized in humans and mice, but M1/M2 profile in cattle has been far less explored. Bos primigenius taurus (taurine) and Bos primigenius indicus (indicine) cattle display contrasting levels of resistance to infection and parasitic diseases such as C57BL/6J and Balb/c murine experimental models of parasite infection outcomes based on genetic background. Thus, we investigated the differential gene expression profile of unstimulated and LPS stimulated monocyte-derived macrophages (MDMs) from Holstein (taurine) and Gir (indicine) breeds using RNA sequencing methodology. For unstimulated MDMs, the contrast between Holstein and Gir breeds identified 163 Differentially Expressed Genes (DEGs) highlighting the higher expression of C-C chemokine receptor type five (CCR5) and BOLA-DQ genes in Gir animals. LPS-stimulated MDMs from Gir and Holstein animals displayed 1,257 DEGs enriched for cell adhesion and inflammatory responses. Gir MDMs cells displayed a higher expression of M1 related genes like Nitric Oxide Synthase 2 (NOS2), Toll like receptor 4 (TLR4), Nuclear factor NF-kappa-B 2 (NFKB2) in addition to higher levels of transcripts for proinflammatory cytokines, chemokines, complement factors and the acute phase protein Serum Amyloid A (SAA). We also showed that gene expression of inflammatory M1 population markers, complement and SAA genes was higher in Gir in buffy coat peripheral cells in addition to nitric oxide concentration in MDMs supernatant and animal serum. Co-expression analyses revealed that Holstein and Gir animals showed different transcriptional signatures in the MDMs response to LPS that impact on cell cycle regulation, leukocyte migration and extracellular matrix organization biological processes. Overall, the results suggest that Gir animals show a natural propensity to generate a more pronounced M1 inflammatory response than Holstein, which might account for a faster immune response favouring resistance to many infection diseases.
Collapse
|