1
|
Changaei M, Azimzadeh Tabrizi Z, Karimi M, Kashfi SA, Koochaki Chahardeh T, Hashemi SM, Soudi S. From powerhouse to modulator: regulating immune system responses through intracellular mitochondrial transfer. Cell Commun Signal 2025; 23:232. [PMID: 40394666 PMCID: PMC12090700 DOI: 10.1186/s12964-025-02237-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
Mitochondria are traditionally known as the cells' powerhouses; however, their roles go far beyond energy suppliers. They are involved in intracellular signaling and thus play a crucial role in shaping cells' destiny and functionality, including immune cells. Mitochondria can be actively exchanged between immune and non-immune cells via mechanisms such as nanotubes and extracellular vesicles. The mitochondria transfer from immune cells to different cells is associated with physiological and pathological processes, including inflammatory disorders, cardiovascular diseases, diabetes, and cancer. On the other hand, mitochondrial transfer from mesenchymal stem cells, bone marrow-derived stem cells, and adipocytes to immune cells significantly affects their functions. Mitochondrial transfer can prevent exhaustion/senescence in immune cells through intracellular signaling pathways and metabolic reprogramming. Thus, it is emerging as a promising therapeutic strategy for immune system diseases, especially those involving inflammation and autoimmune components. Transferring healthy mitochondria into damaged or dysfunctional cells can restore mitochondrial function, which is crucial for cellular energy production, immune regulation, and inflammation control. Also, mitochondrial transfer may enhance the potential of current therapeutic immune cell-based therapies such as CAR-T cell therapy.
Collapse
Affiliation(s)
- Mostafa Changaei
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Azimzadeh Tabrizi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mozhdeh Karimi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Adnan Kashfi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Tina Koochaki Chahardeh
- Department of Basic Sciences, Biology and Health, Faculty of Interdisciplinary Sciences and Technologies, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Elfiky AMI, Canñizares JL, Li J, Li Yim AYF, Verhoeven AJ, Ghiboub M, de Jonge WJ. Carboxylesterase 1 directs the metabolic profile of dendritic cells to a reduced inflammatory phenotype. J Leukoc Biol 2024; 116:1094-1108. [PMID: 38869086 DOI: 10.1093/jleuko/qiae137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 03/17/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
The metabolic profile of dendritic cells (DCs) shapes their phenotype and functions. The carboxylesterase 1 (CES1) enzyme is highly expressed in mononuclear myeloid cells; however, its exact role in DCs is elusive. We used a CES1 inhibitor (WWL113) and genetic overexpression to explore the role of CES1 in DC differentiation in inflammatory models. CES1 expression was analyzed during CD14+ monocytes differentiation to DCs (MoDCs) using quantitative polymerase chain reaction. A CES1 inhibitor (WWL113) was applied during MoDC differentiation. Surface markers, secreted cytokines, lactic acid production, and phagocytic and T cell polarization capacity were analyzed. The transcriptomic and metabolic profiles were assessed with RNA sequencing and mass spectrometry, respectively. Cellular respiration was assessed using seahorse respirometry. Transgenic mice were used to assess the effect of CES1 overexpression in DCs in inflammatory models. CES1 expression peaked early during MoDC differentiation. Pharmacological inhibition of CES1 led to higher expression of CD209, CD86 and MHCII. WWL113 treated MoDCs secreted higher quantities of interleukin (IL)-6, IL-8, tumor necrosis factor, and IL-10 and demonstrated stronger phagocytic ability and a higher capacity to polarize T helper 17 differentiation in an autologous DC-T cell coculture model. Transcriptomic profiling revealed enrichment of multiple inflammatory and metabolic pathways. Functional metabolic analysis showed impaired maximal mitochondrial respiration capacity, increased lactate production, and decreased intracellular amino acids and tricarboxylic acid cycle intermediates. Transgenic human CES1 overexpression in murine DCs generated a less inflammatory phenotype and increased resistance to T cell-mediated colitis. In conclusion, CES1 inhibition directs DC differentiation toward a more inflammatory phenotype that shows a stronger phagocytic capacity and supports T helper 17 skewing. This is associated with a disrupted mitochondrial respiration and amino acid depletion.
Collapse
Affiliation(s)
- Ahmed M I Elfiky
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 69, 1105 BK, Amsterdam, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Jessica López Canñizares
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 69, 1105 BK, Amsterdam, the Netherlands
| | - Jiarong Li
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 69, 1105 BK, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Andrew Y F Li Yim
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 69, 1105 BK, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Emma Children's Hospital, Pediatric Surgery, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Arthur J Verhoeven
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 69, 1105 BK, Amsterdam, the Netherlands
| | - Mohammed Ghiboub
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 69, 1105 BK, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Emma Children's Hospital, Pediatric Surgery, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 69, 1105 BK, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Amsterdam Infection and Immunity Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
- Department of Surgery, University of Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany
| |
Collapse
|
3
|
Wang Q, Yin X, Huang X, Zhang L, Lu H. Impact of mitochondrial dysfunction on the antitumor effects of immune cells. Front Immunol 2024; 15:1428596. [PMID: 39464876 PMCID: PMC11502362 DOI: 10.3389/fimmu.2024.1428596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
Mitochondrial dysfunction, a hallmark of immune cell failure, affects the antitumor effects of immune cells through metabolic reprogramming, fission, fusion, biogenesis, and immune checkpoint signal transduction of mitochondria. According to researchers, restoring damaged mitochondrial function can enhance the efficacy of immune cells. Nevertheless, the mechanism of mitochondrial dysfunction in immune cells in patients with cancer is unclear. In this review, we recapitulate the impact of mitochondrial dysfunction on the antitumor effects of T cells, natural killer cells, dendritic cells, and tumor-associated macrophage and propose that targeting mitochondria can provide new strategies for antitumor therapy.
Collapse
Affiliation(s)
- Quan Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangzhi Yin
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Huang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haijun Lu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Rieder AS, Ramires Júnior OV, Prauchner GRK, Wyse ATS. Effects of methylphenidate on mitochondrial dynamics and bioenergetics in the prefrontal cortex of juvenile rats are sex-dependent. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111057. [PMID: 38880464 DOI: 10.1016/j.pnpbp.2024.111057] [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: 12/07/2023] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
Methylphenidate (MPH) is a central nervous system stimulant drug and a first order prescription in the treatment of Attention Deficit Hyperactivity Disorder (ADHD). Although MPH biochemistry in neurodevelopment is not completely understood, studies showed it alters energy metabolism in rat brains. ADHD prevalence during neurodevelopment is related to males and the investigation has been mainly done in these subjects, therefore, little is known about MPH action in females and, consequently, about sexual dimorphism. In the present study we evaluated markers of mitochondrial dynamics (DRP1 and MFN2, fission and fusion, respectively), biogenesis (mtTFA) and bioenergetics (respiratory chain complexes) in prefrontal cortex of male and female juvenile rats submitted to exposure to MPH to better understand MPH effect during postnatal neurodevelopment. ATP and oxidative stress levels were also evaluated. Wistar rats received intraperitoneal injection of MPH (2.0 mg/kg) or control (saline), once a day, from 15th to 45th day of age. Results showed that MPH increased DRP1 and decreased MFN2, as well as increased mtTFA in prefrontal cortex of male rats. In female, MPH decreased NRF1 and increased Parkin, which are mitochondrial regulatory proteins. Respiratory chain complexes (complex I, SDH, complexes III and IV), ATP production and oxidative stress parameters were altered and shown to be sex-dependent. Taken together, results suggest that chronic MPH exposure at an early age in healthy animals changes mitochondrial dynamics, biogenesis and bioenergetics differently depending on the sex of the subjects.
Collapse
Affiliation(s)
- Alessandra Schmitt Rieder
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | - Osmar Vieira Ramires Júnior
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | - Gustavo Ricardo Krupp Prauchner
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
5
|
Liu Z, Zeinalzadeh Z, Huang T, Han Y, Peng L, Wang D, Zhou Z, Ousmane D, Wang J. Mitochondria-related chemoradiotherapy resistance genes-based machine learning model associated with immune cell infiltration on the prognosis of esophageal cancer and its value in pan-cancer. Transl Oncol 2024; 42:101896. [PMID: 38324960 PMCID: PMC10851222 DOI: 10.1016/j.tranon.2024.101896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024] Open
Abstract
Esophageal cancer, known for its high incidence and low five-year survival rate, poses significant treatment challenges. A key aspect of this challenge is the close link between mitochondria and resistance to chemoradiotherapy (CRT). Currently, there is a scarcity of biomarkers for predicting CRT response and prognosis in esophageal cancer. Our study addresses this gap by developing a prognostic model that incorporates mitochondria-related CRT resistance (MRCRTR) genes, including CTSL, TBL1X, CLN8, MMP1, PDPN, and MRPL37. Survival analysis using Kaplan-Meier curves reveals that patients with high MRCRTR scores have lower survival rates than those with low scores. Utilizing a nomogram, we successfully predict the one-, two-, and three-year overall survival rates for esophageal cancer patients. Cox regression analysis confirms the MRCRTR score as an independent prognostic factor. Furthermore, our single-cell and correlation analyses suggested that MRCRTR genes might influence CRT resistance by modulating the immune microenvironment and impacting angiogenesis. Our pan-cancer analysis also indicates the potential applicability of MRCRTR scores to head and neck squamous cell carcinoma. The validation of these findings, conducted with samples from Xiang-ya Hospital, aligns closely with our bioinformatics results. Our study not only explores the role of MRCRTR genes in predicting the prognosis of esophageal cancer but also enhances the understanding of the interplay between CRT, mitochondria, and patient outcomes.
Collapse
Affiliation(s)
- Ziyu Liu
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China; Ultrapathology (Biomedical Electron Microscopy) Center, Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zahra Zeinalzadeh
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Tao Huang
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Yingying Han
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Lushan Peng
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Dan Wang
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Zongjiang Zhou
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Diabate Ousmane
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Junpu Wang
- Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China; Ultrapathology (Biomedical Electron Microscopy) Center, Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
6
|
Matrella ML, Valletti A, Gigante I, De Rasmo D, Signorile A, Russo S, Lobasso S, Lobraico D, Dibattista M, Pacelli C, Cocco T. High OXPHOS efficiency in RA-FUdr-differentiated SH-SY5Y cells: involvement of cAMP signalling and respiratory supercomplexes. Sci Rep 2024; 14:7411. [PMID: 38548913 PMCID: PMC10978939 DOI: 10.1038/s41598-024-57613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/20/2024] [Indexed: 04/01/2024] Open
Abstract
Neurons are highly dependent on mitochondria to meet their bioenergetic needs and understanding the metabolic changes during the differentiation process is crucial in the neurodegeneration context. Several in vitro approaches have been developed to study neuronal differentiation and bioenergetic changes. The human SH-SY5Y cell line is a widely used cellular model and several differentiation protocols have been developed to induce a neuron-like phenotype including retinoic acid (RA) treatment. In this work we obtained a homogeneous functional population of neuron-like cells by a two-step differentiation protocol in which SH-SY5Y cells were treated with RA plus the mitotic inhibitor 2-deoxy-5-fluorouridine (FUdr). RA-FUdr treatment induced a neuronal phenotype characterized by increased expression of neuronal markers and electrical properties specific to excitable cells. In addition, the RA-FUdr differentiated cells showed an enrichment of long chain and unsaturated fatty acids (FA) in the acyl chain composition of cardiolipin (CL) and the bioenergetic analysis evidences a high coupled and maximal respiration associated with high mitochondrial ATP levels. Our results suggest that the observed high oxidative phosphorylation (OXPHOS) capacity may be related to the activation of the cyclic adenosine monophosphate (cAMP) pathway and the assembly of respiratory supercomplexes (SCs), highlighting the change in mitochondrial phenotype during neuronal differentiation.
Collapse
Affiliation(s)
- Maria Laura Matrella
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Alessio Valletti
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- MASMEC Biomed S.p.A, 70026, Modugno, Italy
| | - Isabella Gigante
- National Institute of Gastroenterology- IRCCS "Saverio De Bellis", Via Turi 27, Castellana Grotte, 70013, Bari, Italy
| | - Domenico De Rasmo
- Bioenergetics and Molecular Biotechnologies, CNR-Institute of Biomembranes, 70124, Bari, Italy
| | - Anna Signorile
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Silvia Russo
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Simona Lobasso
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Donatella Lobraico
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Michele Dibattista
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Italy.
| | - Tiziana Cocco
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy.
| |
Collapse
|
7
|
Adamik J, Munson PV, Maurer DM, Hartmann FJ, Bendall SC, Argüello RJ, Butterfield LH. Immuno-metabolic dendritic cell vaccine signatures associate with overall survival in vaccinated melanoma patients. Nat Commun 2023; 14:7211. [PMID: 37938561 PMCID: PMC10632482 DOI: 10.1038/s41467-023-42881-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
Efficacy of cancer vaccines remains low and mechanistic understanding of antigen presenting cell function in cancer may improve vaccine design and outcomes. Here, we analyze the transcriptomic and immune-metabolic profiles of Dendritic Cells (DCs) from 35 subjects enrolled in a trial of DC vaccines in late-stage melanoma (NCT01622933). Multiple platforms identify metabolism as an important biomarker of DC function and patient overall survival (OS). We demonstrate multiple immune and metabolic gene expression pathway alterations, a functional decrease in OCR/OXPHOS and increase in ECAR/glycolysis in patient vaccines. To dissect molecular mechanisms, we utilize single cell SCENITH functional profiling and show patient clinical outcomes (OS) correlate with DC metabolic profile, and that metabolism is linked to immune phenotype. With single cell metabolic regulome profiling, we show that MCT1 (monocarboxylate transporter-1), a lactate transporter, is increased in patient DCs, as is glucose uptake and lactate secretion. Importantly, pre-vaccination circulating myeloid cells in patients used as precursors for DC vaccine generation are significantly skewed metabolically as are several DC subsets. Together, we demonstrate that the metabolic profile of DC is tightly associated with the immunostimulatory potential of DC vaccines from cancer patients. We link phenotypic and functional metabolic changes to immune signatures that correspond to suppressed DC differentiation.
Collapse
Affiliation(s)
- Juraj Adamik
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, 94129, USA
| | - Paul V Munson
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, 94129, USA
| | - Deena M Maurer
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, 94129, USA
| | - Felix J Hartmann
- Systems Immunology and Single-Cell Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sean C Bendall
- Department of Pathology, Stanford University, Palo Alto, CA, 94304, USA
| | - Rafael J Argüello
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Lisa H Butterfield
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, 94129, USA.
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Zhao Y, Gao C, Liu L, Wang L, Song Z. The development and function of human monocyte-derived dendritic cells regulated by metabolic reprogramming. J Leukoc Biol 2023; 114:212-222. [PMID: 37232942 DOI: 10.1093/jleuko/qiad062] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/15/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Human monocyte-derived dendritic cells (moDCs) that develop from monocytes play a key role in innate inflammatory responses as well as T cell priming. Steady-state moDCs regulate immunogenicity and tolerogenicity by changing metabolic patterns to participate in the body's immune response. Increased glycolytic metabolism after danger signal induction may strengthen moDC immunogenicity, whereas high levels of mitochondrial oxidative phosphorylation were associated with the immaturity and tolerogenicity of moDCs. In this review, we discuss what is currently known about differential metabolic reprogramming of human moDC development and distinct functional properties.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Dermatology, Southwest Hospital, Army Medical University, 30 Gaotanyan Street, District Shapingba, Chongqing, 400038, China
| | - Cuie Gao
- Department of Dermatology, Southwest Hospital, Army Medical University, 30 Gaotanyan Street, District Shapingba, Chongqing, 400038, China
| | - Lu Liu
- Department of Dermatology, Southwest Hospital, Army Medical University, 30 Gaotanyan Street, District Shapingba, Chongqing, 400038, China
| | - Li Wang
- Institute of Immunology, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Zhiqiang Song
- Department of Dermatology, Southwest Hospital, Army Medical University, 30 Gaotanyan Street, District Shapingba, Chongqing, 400038, China
| |
Collapse
|
9
|
Zhang X, Liu J, Cheng Y, Chen K, Chen Y, Zhu H, Li Z, Liu S, Cao X. Metabolic enzyme Suclg2 maintains tolerogenicity of regulatory dendritic cells diffDCs by suppressing Lactb succinylation. J Autoimmun 2023; 138:103048. [PMID: 37216870 DOI: 10.1016/j.jaut.2023.103048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023]
Abstract
Metabolic reprogramming plays a pivotal role in the differentiation and function of immune cells including dendritic cells (DCs). Regulatory DCs can be generated in regional tissue niches like splenic stroma and act as an important part of stromal control of immune response for the maintenance of immune tolerance. However, the metabolic alterations during splenic stroma-driven regulatory DCs differentiation and the metabolic enzyme involved in regulatory DCs function remain poorly understood. By combining metabolomic, transcriptomic, and functional investigations of mature DCs (maDCs) and diffDCs (regulatory DCs differentiated from activated mature DCs through coculturing with splenic stroma), here we identified succinate-CoA ligase subunit beta Suclg2 as a key metabolic enzyme that reprograms the proinflammatory status of mature DCs into a tolerogenic phenotype via preventing NF-κB signaling activation. diffDCs downregulate succinic acid levels and increase the Suclg2 expression along with their differentiation from mature DCs. Suclg2-interference impaired the tolerogenic function of diffDCs in inducing T cell apoptosis and enhanced activation of NF-κB signaling and expression of inflammatory genes CD40, Ccl5, and Il12b in diffDCs. Furthermore, we identified Lactb as a new positive regulator of NF-κB signaling in diffDCs whose succinylation at the lysine 288 residue was inhibited by Suclg2. Our study reveals that the metabolic enzyme Suclg2 is required to maintain the immunoregulatory function of diffDCs, adding mechanistic insights into the metabolic regulation of DC-based immunity and tolerance.
Collapse
Affiliation(s)
- Xiaomin Zhang
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Juan Liu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China.
| | - Yujie Cheng
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Kun Chen
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yali Chen
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Ha Zhu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Zhiqing Li
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Shuxun Liu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China; Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China; Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
10
|
Ning L, Shishi Z, Bo W, Huiqing L. Targeting immunometabolism against acute lung injury. Clin Immunol 2023; 249:109289. [PMID: 36918041 PMCID: PMC10008193 DOI: 10.1016/j.clim.2023.109289] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening conditions triggered by multiple intra- and extra-pulmonary injury factors, characterized by complicated molecular mechanisms and high mortality. Great strides have been made in the field of immunometabolism to clarify the interplay between intracellular metabolism and immune function in the past few years. Emerging evidence unveils the crucial roles of immunometabolism in inflammatory response and ALI. During ALI, both macrophages and lymphocytes undergo robust metabolic reprogramming and discrete epigenetic changes after activated. Apart from providing ATP and biosynthetic precursors, these metabolic cellular reactions and processes in lung also regulate inflammation and immunity.In fact, metabolic reprogramming involving glucose metabolism and fatty acidoxidation (FAO) acts as a double-edged sword in inflammatory response, which not only drives inflammasome activation but also elicits anti-inflammatory response. Additionally, the features and roles of metabolic reprogramming in different immune cells are not exactly the same. Here, we outline the evidence implicating how adverse factors shape immunometabolism in differentiation types of immune cells during ALI and summarize key proteins associated with energy expenditure and metabolic reprogramming. Finally, novel therapeutic targets in metabolic intermediates and enzymes together with current challenges in immunometabolism against ALI were discussed.
Collapse
Affiliation(s)
- Li Ning
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Zou Shishi
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Wang Bo
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China.
| | - Lin Huiqing
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China.
| |
Collapse
|
11
|
Alba G, Dakhaoui H, Santa-Maria C, Palomares F, Cejudo-Guillen M, Geniz I, Sobrino F, Montserrat-de la Paz S, Lopez-Enriquez S. Nutraceuticals as Potential Therapeutic Modulators in Immunometabolism. Nutrients 2023; 15:411. [PMID: 36678282 PMCID: PMC9865834 DOI: 10.3390/nu15020411] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/14/2023] Open
Abstract
Nutraceuticals act as cellular and functional modulators, contributing to the homeostasis of physiological processes. In an inflammatory microenvironment, these functional foods can interact with the immune system by modulating or balancing the exacerbated proinflammatory response. In this process, immune cells, such as antigen-presenting cells (APCs), identify danger signals and, after interacting with T lymphocytes, induce a specific effector response. Moreover, this conditions their change of state with phenotypical and functional modifications from the resting state to the activated and effector state, supposing an increase in their energy requirements that affect their intracellular metabolism, with each immune cell showing a unique metabolic signature. Thus, nutraceuticals, such as polyphenols, vitamins, fatty acids, and sulforaphane, represent an active option to use therapeutically for health or the prevention of different pathologies, including obesity, metabolic syndrome, and diabetes. To regulate the inflammation associated with these pathologies, intervention in metabolic pathways through the modulation of metabolic energy with nutraceuticals is an attractive strategy that allows inducing important changes in cellular properties. Thus, we provide an overview of the link between metabolism, immune function, and nutraceuticals in chronic inflammatory processes associated with obesity and diabetes, paying particular attention to nutritional effects on APC and T cell immunometabolism, as well as the mechanisms required in the change in energetic pathways involved after their activation.
Collapse
Affiliation(s)
- Gonzalo Alba
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville. Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Hala Dakhaoui
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville. Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Consuelo Santa-Maria
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, 41012 Seville, Spain
| | - Francisca Palomares
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville. Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Marta Cejudo-Guillen
- Department of Pharmacology, Pediatry, and Radiology, School of Medicine, University of Seville. Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Isabel Geniz
- Distrito Sanitario Seville Norte y Aljarafe, Servicio Andaluz de Salud, 41008 Seville, Spain
| | - Francisco Sobrino
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville. Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville. Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Soledad Lopez-Enriquez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville. Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| |
Collapse
|
12
|
Tsymbalyuk O, Gerzanich V, Simard JM, Rathinam CV. Traumatic brain injury alters dendritic cell differentiation and distribution in lymphoid and non-lymphoid organs. J Neuroinflammation 2022; 19:238. [PMID: 36183126 PMCID: PMC9526328 DOI: 10.1186/s12974-022-02609-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 09/24/2022] [Indexed: 11/10/2022] Open
Abstract
Background Pathophysiological consequences of traumatic brain injury (TBI) mediated secondary injury remain incompletely understood. In particular, the impact of TBI on the differentiation and maintenance of dendritic cells (DCs), which are regarded as the most professional antigen presenting cells of the immune system, remains completely unknown. Here, we report that DC-differentiation, maintenance and functions are altered on day 3 and day 7 after TBI. Methods Long bones, spleen, peripheral lymph nodes (pLNs), mesenteric lymph nodes (mLNs), liver, lungs, skin and blood were collected from mice with either moderate-level cortical impact (CCI) or sham on day 1, day 3 or day 7 after TBI. Bone marrow cells were isolated from the tibias and femurs of hind limb through flushing. Tissues were digested with Collagenase-D and DNase I. Skin biopsies were digested in the presence of liberase + DNase I. Single cell suspensions were made, red blood cells were lysed with Ammonium chloride (Stem Cell Technology) and subsequently filtered using a 70 μM nylon mesh. DC subsets of the tissues and DC progenitors of the BM were identified through 10-color flow cytometry-based immunophenotyping studies. Intracellular reactive oxygen species (ROS) were identified through H2DCFDA staining. Results Our studies identify that; (1) frequencies and absolute numbers of DCs in the spleen and BM are altered on day 3 and day 7 after TBI; (2) surface expression of key molecules involved in antigen presentation of DCs were affected on day 3 and day 7 after TBI; (3) distribution and functions of tissue-specific DC subsets of both circulatory and lymphatic systems were imbalanced following TBI; (4) early differentiation program of DCs, especially the commitment of hematopoietic stem cells to common DC progenitors (CDPs), were deregulated after TBI; and (5) intracellular ROS levels were reduced in DC progenitors and differentiated DCs on day 3 and day 7 after TBI. Conclusions Our data demonstrate, for the first time, that TBI affects the distribution pattern of DCs and induces an imbalance among DC subsets in both lymphoid and non-lymphoid organs. In addition, the current study demonstrates that TBI results in reduced levels of ROS in DCs on day 3 and day 7 after TBI, which may explain altered DC differentiation paradigm following TBI. A deeper understanding on the molecular mechanisms that contribute to DC defects following TBI would be essential and beneficial in treating infections in patients with acute central nervous system (CNS) injuries, such as TBI, stroke and spinal cord injury.
Collapse
Affiliation(s)
- Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA.,Research Service, Veterans Affairs Maryland Health Care System, MD, Baltimore, USA.,Department of Pathology, University of Maryland School of Medicine, MD, Baltimore, USA.,Department of Physiology, University of Maryland School of Medicine, MD, Baltimore, USA
| | - Chozha Vendan Rathinam
- Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD, 21201, USA. .,Center for Stem Cell and Regenerative Medicine, University of Maryland School of Medicine, MD, 21201, Baltimore, USA.
| |
Collapse
|
13
|
Distinct metabolic states guide maturation of inflammatory and tolerogenic dendritic cells. Nat Commun 2022; 13:5184. [PMID: 36056019 PMCID: PMC9440236 DOI: 10.1038/s41467-022-32849-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 08/20/2022] [Indexed: 12/17/2022] Open
Abstract
Cellular metabolism underpins immune cell functionality, yet our understanding of metabolic influences in human dendritic cell biology and their ability to orchestrate immune responses is poorly developed. Here, we map single-cell metabolic states and immune profiles of inflammatory and tolerogenic monocytic dendritic cells using recently developed multiparametric approaches. Single-cell metabolic pathway activation scores reveal simultaneous engagement of multiple metabolic pathways in distinct monocytic dendritic cell differentiation stages. GM-CSF/IL4-induce rapid reprogramming of glycolytic monocytes and transient co-activation of mitochondrial pathways followed by TLR4-dependent maturation of dendritic cells. Skewing of the mTOR:AMPK phosphorylation balance and upregulation of OXPHOS, glycolytic and fatty acid oxidation metabolism underpin metabolic hyperactivity and an immunosuppressive phenotype of tolerogenic dendritic cells, which exhibit maturation-resistance and a de-differentiated immune phenotype marked by unique immunoregulatory receptor signatures. This single-cell dataset provides important insights into metabolic pathways impacting the immune profiles of human dendritic cells. Assessing metabolic activity within single cells rather than at a population level has a number of advantages. Here, the authors use a flow and mass cytometry based approach that assess the metabolic differences between populations of human immune stimulatory and tolerogenic dendritic cells.
Collapse
|
14
|
Liu J, Zhou G, Wang X, Liu D. Metabolic reprogramming consequences of sepsis: adaptations and contradictions. Cell Mol Life Sci 2022; 79:456. [PMID: 35904600 PMCID: PMC9336160 DOI: 10.1007/s00018-022-04490-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 12/19/2022]
Abstract
During sepsis, the importance of alterations in cell metabolism is underappreciated. The cellular metabolism, which has a variable metabolic profile in different cells and disease stages, is largely responsible for the immune imbalance and organ failure associated with sepsis. Metabolic reprogramming, in which glycolysis replaces OXPHOS as the main energy-producing pathway, is both a requirement for immune cell activation and a cause of immunosuppression. Meanwhile, the metabolites produced by OXPHOS and glycolysis can act as signaling molecules to control the immune response during sepsis. Sepsis-induced "energy shortage" leads to stagnated cell function and even organ dysfunction. Metabolic reprogramming can alleviate the energy crisis to some extent, enhance host tolerance to maintain cell survival functions, and ultimately increase the adaptation of cells during sepsis. However, a switch from glycolysis to OXPHOS is essential for restoring cell function. This review summarized the crosstalk between metabolic reprogramming and immune cell activity as well as organ function during sepsis, discussed the benefits and drawbacks of metabolic reprogramming to show the contradictions of metabolic reprogramming during sepsis, and assessed the feasibility of treating sepsis through targeted metabolism. Using metabolic reprogramming to achieve metabolic homeostasis could be a viable therapy option for sepsis.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Cheng District, Beijing, 100730 China
| | - Gaosheng Zhou
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Cheng District, Beijing, 100730 China
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Cheng District, Beijing, 100730 China
| | - Dawei Liu
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Cheng District, Beijing, 100730 China
| |
Collapse
|
15
|
Wilson KR, Gressier E, McConville MJ, Bedoui S. Microbial Metabolites in the Maturation and Activation of Dendritic Cells and Their Relevance for Respiratory Immunity. Front Immunol 2022; 13:897462. [PMID: 35880171 PMCID: PMC9307905 DOI: 10.3389/fimmu.2022.897462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
The respiratory tract is a gateway for viruses and bacteria from the external environment to invade the human body. Critical to the protection against these invaders are dendritic cells (DCs) - a group of highly specialized myeloid cells that monitors the lung microenvironment and relays contextual and antigenic information to T cells. Following the recognition of danger signals and/or pathogen molecular associated patterns in the lungs, DCs undergo activation. This process arms DCs with the unique ability to induce the proliferation and differentiation of T cells responding to matching antigen in complex with MHC molecules. Depending on how DCs interact with T cells, the ensuing T cell response can be tolerogenic or immunogenic and as such, the susceptibility and severity of respiratory infections is influenced by the signals DCs receive, integrate, and then convey to T cells. It is becoming increasingly clear that these facets of DC biology are heavily influenced by the cellular components and metabolites produced by the lung and gut microbiota. In this review, we discuss the roles of different DC subsets in respiratory infections and outline how microbial metabolites impact the development, propensity for activation and subsequent activation of DCs. In particular, we highlight these concepts in the context of respiratory immunity.
Collapse
Affiliation(s)
- Kayla R. Wilson
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Kayla R. Wilson,
| | - Elise Gressier
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Malcolm J. McConville
- Department of Biochemistry and Pharmacology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, VIC, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
16
|
Møller SH, Wang L, Ho PC. Metabolic programming in dendritic cells tailors immune responses and homeostasis. Cell Mol Immunol 2022; 19:370-383. [PMID: 34413487 PMCID: PMC8891341 DOI: 10.1038/s41423-021-00753-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023] Open
Abstract
It is being increasingly acknowledged that immune cells depend on certain metabolic traits to perform their functions and that the extracellular environment can influence cell metabolism and vice versa. Dendritic cell (DC) subsets traffic through highly diverse environments from the bone marrow, where they develop, to the various peripheral tissues, where they differentiate and capture antigens, before they migrate to the lymph node to present antigens and prime T cells. It is plausible that DC subsets modulate their stimulatory abilities in response to unique metabolic programming. The metabolic requirements of DCs are just recently being discovered, and subset- and context-specific metabolic phenotypes in DCs are highly intertwined with DC functions. In this review, we present the current knowledge on the intrinsic and extrinsic determinants of DC metabolism, how they regulate DC function with examples from tumor biology and in interaction with the microbiota, and discuss how this can be applied therapeutically.
Collapse
Affiliation(s)
- Sofie Hedlund Møller
- grid.9851.50000 0001 2165 4204Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland ,grid.9851.50000 0001 2165 4204Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Limei Wang
- grid.9851.50000 0001 2165 4204Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland ,grid.9851.50000 0001 2165 4204Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Ping-Chih Ho
- grid.9851.50000 0001 2165 4204Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland ,grid.9851.50000 0001 2165 4204Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
17
|
Chen H, Fan W, He H, Huang F. PGC-1: a key regulator in bone homeostasis. J Bone Miner Metab 2022; 40:1-8. [PMID: 34424416 DOI: 10.1007/s00774-021-01263-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
Peroxisome proliferator-activated receptor-γ coactivator-1 (PGC-1) is an inducible co-regulator of nuclear receptors and is involved in a wide variety of biological responses. As the master regulators of mitochondrial biogenesis and function, PGC-1α and PGC-1β have been reported to play key roles in bone metabolism. They can be rapidly induced under conditions of increased metabolic activities, such as osteoblastogenesis and osteoclastogenesis, to fulfill greater energy demand or facilitate other biochemical reactions. PGC-1α and PGC-1β have both overlapping and distinct functions with each other among their target organs. In bone homeostasis, PGC-1α and PGC-1β promote the expression of genes required for mitochondrial biogenesis via coactivator interactions with key transcription factors, respectively regulating osteoblastogenesis and osteoclastogenesis. Here, we review the current understanding of how PGC-1α and PGC-1β affect osteoblastogenesis and osteoclastogenesis, how these two PGC-1 coactivators are regulated in bone homeostasis, and how we can translate these findings into therapeutic potential for bone metabolic diseases.
Collapse
Affiliation(s)
- Haoling Chen
- Hospital of Stomatology, Sun Yat-Sen University, 56 Lingyuan Xi Road, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Wenguo Fan
- Hospital of Stomatology, Sun Yat-Sen University, 56 Lingyuan Xi Road, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China.
| | - Fang Huang
- Hospital of Stomatology, Sun Yat-Sen University, 56 Lingyuan Xi Road, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China.
| |
Collapse
|
18
|
Neuro-immune-metabolism: The tripod system of homeostasis. Immunol Lett 2021; 240:77-97. [PMID: 34655659 DOI: 10.1016/j.imlet.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 11/20/2022]
Abstract
Homeostatic regulation of cellular and molecular processes is essential for the efficient physiological functioning of body organs. It requires an intricate balance of several networks throughout the body, most notable being the nervous, immune and metabolic systems. Several studies have reported the interactions between neuro-immune, immune-metabolic and neuro-metabolic pathways. Current review aims to integrate the information and show that neuro, immune and metabolic systems form the triumvirate of homeostasis. It focuses on the cellular and molecular interactions occurring in the extremities and intestine, which are innervated by the peripheral nervous system and for the intestine in particular the enteric nervous system. While the interdependence of neuro-immune-metabolic pathways provides a fallback mechanism in case of disruption of homeostasis, in chronic pathologies of continued disequilibrium, the collapse of one system spreads to the other interacting networks as well. Current review illustrates this domino-effect using diabetes as the main example. Together, this review attempts to provide a holistic picture of the integrated network of neuro-immune-metabolism and attempts to broaden the outlook when devising a scientific study or a treatment strategy.
Collapse
|
19
|
Cervantes‐Silva MP, Cox SL, Curtis AM. Alterations in mitochondrial morphology as a key driver of immunity and host defence. EMBO Rep 2021; 22:e53086. [PMID: 34337844 PMCID: PMC8447557 DOI: 10.15252/embr.202153086] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/09/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are dynamic organelles whose architecture changes depending on the cell's energy requirements and other signalling events. These structural changes are collectively known as mitochondrial dynamics. Mitochondrial dynamics are crucial for cellular functions such as differentiation, energy production and cell death. Importantly, it has become clear in recent years that mitochondrial dynamics are a critical control point for immune cell function. Mitochondrial remodelling allows quiescent immune cells to rapidly change their metabolism and become activated, producing mediators, such as cytokines, chemokines and even metabolites to execute an effective immune response. The importance of mitochondrial dynamics in immunity is evident, as numerous pathogens have evolved mechanisms to manipulate host cell mitochondrial remodelling in order to promote their own survival. In this review, we comprehensively address the roles of mitochondrial dynamics in immune cell function, along with modulation of host cell mitochondrial morphology during viral and bacterial infections to facilitate either pathogen survival or host immunity. We also speculate on what the future may hold in terms of therapies targeting mitochondrial morphology for bacterial and viral control.
Collapse
Affiliation(s)
- Mariana P Cervantes‐Silva
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| | - Shannon L Cox
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| | - Annie M Curtis
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
20
|
Patel J, Maddukuri S, Li Y, Bax C, Werth VP. Highly Multiplexed Mass Cytometry Identifies the Immunophenotype in the Skin of Dermatomyositis. J Invest Dermatol 2021; 141:2151-2160. [PMID: 33766508 PMCID: PMC8384654 DOI: 10.1016/j.jid.2021.02.748] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/31/2021] [Accepted: 02/17/2021] [Indexed: 01/06/2023]
Abstract
Dermatomyositis (DM) is a rare, systemic autoimmune disease that most frequently affects the skin, muscles, and lungs. The inflammatory infiltrate in the skin has not been fully characterized, and, in this study, we took a single-cell, unbiased approach using imaging mass cytometry. Substantial monocyte‒macrophage diversity was observed, with the CD14+ population correlating positively with Cutaneous Dermatomyositis Disease Area and Severity Index scores (P = 0.031). The T-cell compartment revealed CD4+ T, CD8+ T, and FOXP3+ T cells. Activated (CD69+) circulating memory T cells correlated positively with Cutaneous Dermatomyositis Disease Area and Severity Index scores (P = 0.0268). IFN-β protein was highly upregulated in the T-cell, macrophage, dendritic cell, and endothelial cell populations of DM skin. Myeloid dendritic cells expressed phosphorylated peroxisome proliferator‒activated receptor γ, phosphorylated IRF3, IL-4, and IL-31, and their quantity correlated with itch as measured in Skindex-29. Plasmacytoid dendritic cells colocalized with IFN-γ in addition to the known colocalization with IFN-β. Nuclear phosphorylated peroxisome proliferator‒activated receptor γ expression was found in the DM endothelium. Imaging mass cytometry allows us to characterize single cells in the immune cell population and identify upregulated cytokines and inflammatory pathways in DM. These findings have important implications for the development of future targeted therapies for DM.
Collapse
Affiliation(s)
- Jay Patel
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, USA; Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Spandana Maddukuri
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, USA; Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yubin Li
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, USA; Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christina Bax
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, USA; Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Victoria P Werth
- Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania, USA; Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
21
|
Zhang L, Xia H, Xia K, Liu X, Zhang X, Dai J, Zeng Z, Jia Y. Selenium Regulation of the Immune Function of Dendritic Cells in Mice Through the ERK, Akt and RhoA/ROCK Pathways. Biol Trace Elem Res 2021; 199:3360-3370. [PMID: 33107016 DOI: 10.1007/s12011-020-02449-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/18/2020] [Indexed: 10/23/2022]
Abstract
Selenium levels can regulate the function of T cells, macrophages, B cells, natural killer cells and other immune cells. However, the effect of selenium on the immune function of dendritic cells (DCs) isolated from selenium-supplemented mice is unknown. In this study, C57BL/6J mice were randomly divided into three groups and fed diets containing low (0.08 ppm), medium (0.25 ppm) or high (1 ppm) selenium levels for 8 weeks. Immature (imDCs) and mature (mDCs) dendritic cells were then isolated from the bone marrow. Next, the migration, phagocytic capacity and mixed lymphocyte reaction (MLR) for imDCs and mDCs were detected by transwell and flow cytometry. The levels of C-C chemokine receptor type 7 (CCR7), major histocompatibility complex II (MHCII) and reactive oxygen species (ROS) were assayed by flow cytometry. F-actin and superoxide dismutase (SOD) activity was detected by fluorescence microscopy and SOD assay kit, respectively. In addition, the extracellular signal-regulated kinase (ERK), Akt, Ras homolog gene family member A/Rho-associated protein kinase (RhoA/ROCK) signalling, selenoprotein K (SELENOK) and glutathione peroxidase 1 (GPX1) levels were measured by western blot analysis. The results indicated that selenium deficiency enhanced the migration of imDCs by ROS and SELENOK-mediated ERK, Akt and RhoA/ROCK pathways but impaired the antigen uptake of imDCs. Although a high selenium level inhibited the migration of imDCs, it had no effect on phagocytic capacity. For mDCs, low selenium levels impaired free migration, and high levels inhibited the chemotactic migration involved in F-actin and CCR7, respectively. Low and high selenium levels impaired the MLR by inhibiting MHCII surface localisation, which might be related to ROS- and SELENOK-mediated ERK, Akt and RhoA/ROCK signalling pathways. In summary, selenium may regulate the immune function of mouse DCs through the ROS- and SELENOK-mediated ERK, Akt and RhoA/ROCK signalling.
Collapse
Affiliation(s)
- Liangliang Zhang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Huan Xia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Kaide Xia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xianmei Liu
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xin Zhang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jie Dai
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhu Zeng
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
- School of Basic Medical Science, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yi Jia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China.
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
22
|
Chen JY, Zhou JK, Pan W. Immunometabolism: Towards a Better Understanding the Mechanism of Parasitic Infection and Immunity. Front Immunol 2021; 12:661241. [PMID: 34122419 PMCID: PMC8191844 DOI: 10.3389/fimmu.2021.661241] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/13/2021] [Indexed: 12/26/2022] Open
Abstract
As a relatively successful pathogen, several parasites can establish long-term infection in host. This “harmonious symbiosis” status relies on the “precise” manipulation of host immunity and metabolism, however, the underlying mechanism is still largely elusive. Immunometabolism is an emerging crossed subject in recent years. It mainly discusses the regulatory mechanism of metabolic changes on reprogramming the key transcriptional and post-transcriptional events related to immune cell activation and effect, which provides a novel insight for understanding how parasites regulate the infection and immunity in hosts. The present study reviewed the current research progress on metabolic reprogramming mechanism exploited by parasites to modulate the function in various immune cells, highlighting the future exploitation of key metabolites or metabolic events to clarify the underlying mechanism of anti-parasite immunity and design novel intervention strategies against parasitic infection.
Collapse
Affiliation(s)
- Jing-Yue Chen
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Ji-Kai Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
23
|
Xia H, Zhang L, Dai J, Liu X, Zhang X, Zeng Z, Jia Y. Effect of Selenium and Peroxynitrite on Immune Function of Immature Dendritic Cells in Humans. Med Sci Monit 2021; 27:e929004. [PMID: 33684094 PMCID: PMC7953518 DOI: 10.12659/msm.929004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Selenium and peroxynitrite are known to support the growth and activity of immune cells, including T cells, B cells and macrophages. However, the role of these factors in the immune function of human immature dendritic cells (imDCs) is not clear. Material/Methods Monocytes from a mixture of blood samples were isolated using Ficoll density gradient centrifugation and purified with immunomagnetic beads before being induced into imDCs. Cells then either received no treatment (control group), or treatment with sodium selenite (Na2SeO3, Se), 3-morpholinosydnonimine (SIN1, which decomposes into peroxynitrite), or Se+SIN1. Cell viability, migration, and antiphagocytic abilities, oxidative stress, and protein expression of extracellular signal-regulated kinases (ERK) and MMP2 were assessed using a CCK8 assay, cell counter and flow cytometry, microplate spectrophotometer, and Western blot analysis, respectively. Results Viability of imDCs was unaffected by 0.1 μmol/L of Na2SeO3, although 1 mmol/L of SIN1 decreased it significantly (P<0.05). Chemotactic migration and antiphagocytic abilities were inhibited and enhanced, respectively, by treatment with Na2SeO3 and SIN1 (P<0.05). Activities of superoxide dismutase and glutathione peroxidase were increased by Na2SeO3 and Se+SIN1 (P<0.001). Glutathione content decreased with exposure to Na2SeO3 and SIN1 (P<0.05), but increased after treatment with Se+SIN1 (P<0.05). Levels of reactive oxygen species only increased with SIN1 treatment (P<0.05). Treatment with Na2SeO3, SIN1 and Se+SIN1 increased ERK phosphorylation and decreased MMP2 protein expression (P<0.05). Conclusions Selenium and peroxynitrite can influence immune function in imDCs by regulating levels of reactive oxygen species or glutathione to activate ERK and promote antigen phagocytosis, as well as by decreasing MMP2 expression to inhibit chemotactic migration.
Collapse
Affiliation(s)
- Huan Xia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Liangliang Zhang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Jie Dai
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Xianmei Liu
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Xin Zhang
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland).,Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Zhu Zeng
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Basic Medical Science, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| | - Yi Jia
- Immune Cells and Antibody Engineering Research Center of Guizhou Province/Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland).,School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| |
Collapse
|
24
|
Minarrieta L, Velasquez LN, Sparwasser T, Berod L. Dendritic cell metabolism: moving beyond in vitro-culture-generated paradigms. Curr Opin Biotechnol 2021; 68:202-212. [PMID: 33517147 DOI: 10.1016/j.copbio.2020.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/25/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) are key orchestrators of immunity and tolerance. It has become evident that DC function can be influenced by cellular metabolic programs. However, conclusions from early metabolic studies using in vitro GM-CSF DC cultures fail to correlate with bona fide DC populations. Here, we discuss the existing paradigms in the DC metabolism field, focusing on the limitations of the models utilized. Furthermore, we introduce alternative models to generate DCs in vitro that better emulate DCs found in vivo. Finally, we highlight new techniques to evaluate DC metabolism at the single-cell level. The combination of these two strategies could help advance the DC metabolism field towards a more physiological understanding, which is crucial for the development of effective DC-based therapies.
Collapse
Affiliation(s)
- Lucía Minarrieta
- Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Lis Noelia Velasquez
- Institute of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luciana Berod
- Institute of Molecular Medicine, University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| |
Collapse
|
25
|
Soto‐Heredero G, Gómez de las Heras MM, Gabandé‐Rodríguez E, Oller J, Mittelbrunn M. Glycolysis - a key player in the inflammatory response. FEBS J 2020; 287:3350-3369. [PMID: 32255251 PMCID: PMC7496292 DOI: 10.1111/febs.15327] [Citation(s) in RCA: 342] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/06/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Abstract
The inflammatory response involves the activation of several cell types to fight insults caused by a plethora of agents, and to maintain the tissue homoeostasis. On the one hand, cells involved in the pro-inflammatory response, such as inflammatory M1 macrophages, Th1 and Th17 lymphocytes or activated microglia, must rapidly provide energy to fuel inflammation, which is essentially accomplished by glycolysis and high lactate production. On the other hand, regulatory T cells or M2 macrophages, which are involved in immune regulation and resolution of inflammation, preferentially use fatty acid oxidation through the TCA cycle as a main source for energy production. Here, we discuss the impact of glycolytic metabolism at the different steps of the inflammatory response. Finally, we review a wide variety of molecular mechanisms which could explain the relationship between glycolytic metabolites and the pro-inflammatory phenotype, including signalling events, epigenetic remodelling, post-transcriptional regulation and post-translational modifications. Inflammatory processes are a common feature of many age-associated diseases, such as cardiovascular and neurodegenerative disorders. The finding that immunometabolism could be a master regulator of inflammation broadens the avenue for treating inflammation-related pathologies through the manipulation of the vascular and immune cell metabolism.
Collapse
Affiliation(s)
- Gonzalo Soto‐Heredero
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)MadridSpain
| | - Manuel M. Gómez de las Heras
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
| | - Enrique Gabandé‐Rodríguez
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)MadridSpain
| | - Jorge Oller
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)MadridSpain
| | - María Mittelbrunn
- Immunometabolism and Inflammation LaboratoryCellular Communication & Inflammation UnitCentro de Biología Molecular Severo OchoaMadridSpain
- Department of Molecular BiologyFaculty of SciencesUniversidad Autónoma de Madrid (UAM)Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12)MadridSpain
| |
Collapse
|
26
|
Kotsafti A, Scarpa M, Castagliuolo I, Scarpa M. Reactive Oxygen Species and Antitumor Immunity-From Surveillance to Evasion. Cancers (Basel) 2020; 12:E1748. [PMID: 32630174 PMCID: PMC7409327 DOI: 10.3390/cancers12071748] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 12/14/2022] Open
Abstract
The immune system is a crucial regulator of tumor biology with the capacity to support or inhibit cancer development, growth, invasion and metastasis. Emerging evidence show that reactive oxygen species (ROS) are not only mediators of oxidative stress but also players of immune regulation in tumor development. This review intends to discuss the mechanism by which ROS can affect the anti-tumor immune response, with particular emphasis on their role on cancer antigenicity, immunogenicity and shaping of the tumor immune microenvironment. Given the complex role that ROS play in the dynamics of cancer-immune cell interaction, further investigation is needed for the development of effective strategies combining ROS manipulation and immunotherapies for cancer treatment.
Collapse
Affiliation(s)
- Andromachi Kotsafti
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy;
| | - Marco Scarpa
- General Surgery Unit, Azienda Ospedaliera di Padova, 35128 Padua, Italy;
| | | | - Melania Scarpa
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy;
| |
Collapse
|
27
|
Schaupp L, Muth S, Rogell L, Kofoed-Branzk M, Melchior F, Lienenklaus S, Ganal-Vonarburg SC, Klein M, Guendel F, Hain T, Schütze K, Grundmann U, Schmitt V, Dorsch M, Spanier J, Larsen PK, Schwanz T, Jäckel S, Reinhardt C, Bopp T, Danckwardt S, Mahnke K, Heinz GA, Mashreghi MF, Durek P, Kalinke U, Kretz O, Huber TB, Weiss S, Wilhelm C, Macpherson AJ, Schild H, Diefenbach A, Probst HC. Microbiota-Induced Type I Interferons Instruct a Poised Basal State of Dendritic Cells. Cell 2020; 181:1080-1096.e19. [PMID: 32380006 DOI: 10.1016/j.cell.2020.04.022] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/31/2019] [Accepted: 04/14/2020] [Indexed: 12/12/2022]
Abstract
Environmental signals shape host physiology and fitness. Microbiota-derived cues are required to program conventional dendritic cells (cDCs) during the steady state so that they can promptly respond and initiate adaptive immune responses when encountering pathogens. However, the molecular underpinnings of microbiota-guided instructive programs are not well understood. Here, we report that the indigenous microbiota controls constitutive production of type I interferons (IFN-I) by plasmacytoid DCs. Using genome-wide analysis of transcriptional and epigenetic regulomes of cDCs from germ-free and IFN-I receptor (IFNAR)-deficient mice, we found that tonic IFNAR signaling instructs a specific epigenomic and metabolic basal state that poises cDCs for future pathogen combat. However, such beneficial biological function comes with a trade-off. Instructed cDCs can prime T cell responses against harmless peripheral antigens when removing roadblocks of peripheral tolerance. Our data provide fresh insights into the evolutionary trade-offs that come with successful adaptation of vertebrates to their microbial environment.
Collapse
Affiliation(s)
- Laura Schaupp
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany; Institute for Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
| | - Sabine Muth
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Leif Rogell
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany; Max-Planck-Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Michael Kofoed-Branzk
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany; Max-Planck-Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108 Freiburg, Germany
| | - Felix Melchior
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stefan Lienenklaus
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany; Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Stephanie C Ganal-Vonarburg
- Department for BioMedical Research (DBMR), University Clinic for Visceral Surgery and Medicine, Inselspital, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
| | - Matthias Klein
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Fabian Guendel
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Tobias Hain
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Kristian Schütze
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Ulrike Grundmann
- Institute for Medical Microbiology and Hygiene, University of Freiburg Medical Center, Hermann-Herder-Str. 11, 79104 Freiburg, Germany
| | - Vanessa Schmitt
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Martina Dorsch
- Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Pia-Katharina Larsen
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Thomas Schwanz
- Institute of Medical Microbiology and Hygiene, University Medical Center Mainz, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany
| | - Sven Jäckel
- Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Tobias Bopp
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; University Cancer Center Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; German Cancer Consortium (DKTK)
| | - Sven Danckwardt
- Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Posttranscriptional Gene Regulation, Cancer Research and Experimental Hemostasis, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Institute for Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Karsten Mahnke
- Department of Dermatology, Ruprecht-Karls-University Heidelberg, D-69120 Heidelberg, Germany
| | - Gitta Anne Heinz
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Pawel Durek
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany; Department for Neuroanatomy, Anatomy and Cell Biology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Siegfried Weiss
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Andrew J Macpherson
- Department for BioMedical Research (DBMR), University Clinic for Visceral Surgery and Medicine, Inselspital, University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
| | - Hansjörg Schild
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; Helmholtz Institute Translational Oncology, Obere Zahlbacher Straße 63, 55131 Mainz, Germany.
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10178 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany.
| | - Hans Christian Probst
- Institute of Immunology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Centre for Immunotherapy, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.
| |
Collapse
|
28
|
Kumar R, Singh P, Kolloli A, Shi L, Bushkin Y, Tyagi S, Subbian S. Immunometabolism of Phagocytes During Mycobacterium tuberculosis Infection. Front Mol Biosci 2019; 6:105. [PMID: 31681793 PMCID: PMC6803600 DOI: 10.3389/fmolb.2019.00105] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/26/2019] [Indexed: 12/18/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) remains as a leading killer among infectious diseases worldwide. The nature of the host immune response dictates whether the initial Mtb infection is cleared or progresses toward active disease, and is ultimately determined by intricate host-pathogen interactions that are yet to be fully understood. The early immune response to infection is mediated by innate immune cells, including macrophages and neutrophils that can phagocytose Mtb and mount an antimicrobial response. However, Mtb can exploit these innate immune cells for its survival and dissemination. Recently, it has become clear that the immune response and metabolic remodeling are interconnected, which is highlighted by the rapid evolution of the interdisciplinary field of immunometabolism. It has been proposed that the net outcome to Mtb infection—clearance or chronic disease—is likely a result of combined immunologic and metabolic activities of the immune cells. Indeed, host cells activated by Mtb infection have strikingly different metabolic requirements than naïve/non-infected cells. Macrophages activated by Mtb-derived molecules or upon phagocytosis acquire a phenotype similar to M1 with elevated production of pro-inflammatory molecules and rely on glycolysis and pentose phosphate pathway to meet their bioenergetic and metabolic requirements. In these macrophages, oxidative phosphorylation and fatty acid oxidation are dampened. However, the non-infected/naive, M2-type macrophages are anti-inflammatory and derive their energy from oxidative phosphorylation and fatty acid oxidation. Similar metabolic adaptations also occur in other phagocytes, including dendritic cells, neutrophils upon Mtb infection. This metabolic reprogramming of innate immune cells during Mtb infection can differentially regulate their effector functions, such as the production of cytokines and chemokines, and antimicrobial response, all of which can ultimately determine the outcome of Mtb-host interactions within the granulomas. In this review, we describe key immune cells bolstering host innate response and discuss the metabolic reprogramming in these phagocytes during Mtb infection. We focused on the major phagocytes, including macrophages, dendritic cells and neutrophils and the key regulators involved in metabolic reprogramming, such as hypoxia-inducible factor-1, mammalian target of rapamycin, the cellular myelocytomatosis, peroxisome proliferator-activator receptors, sirtuins, arginases, inducible nitric acid synthase and sphingolipids.
Collapse
Affiliation(s)
- Ranjeet Kumar
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Pooja Singh
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Afsal Kolloli
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Lanbo Shi
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Yuri Bushkin
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Sanjay Tyagi
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
29
|
Breda CNDS, Davanzo GG, Basso PJ, Saraiva Câmara NO, Moraes-Vieira PMM. Mitochondria as central hub of the immune system. Redox Biol 2019; 26:101255. [PMID: 31247505 PMCID: PMC6598836 DOI: 10.1016/j.redox.2019.101255] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/01/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023] Open
Abstract
Nearly 130 years after the first insights into the existence of mitochondria, new rolesassociated with these organelles continue to emerge. As essential hubs that dictate cell fate, mitochondria integrate cell physiology, signaling pathways and metabolism. Thus, recent research has focused on understanding how these multifaceted functions can be used to improve inflammatory responses and prevent cellular dysfunction. Here, we describe the role of mitochondria on the development and function of immune cells, highlighting metabolic aspects and pointing out some metabolic- independent features of mitochondria that sustain cell function.
Collapse
Affiliation(s)
- Cristiane Naffah de Souza Breda
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Gustavo Gastão Davanzo
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Paulo José Basso
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | - Pedro Manoel Mendes Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil.
| |
Collapse
|
30
|
Wculek SK, Khouili SC, Priego E, Heras-Murillo I, Sancho D. Metabolic Control of Dendritic Cell Functions: Digesting Information. Front Immunol 2019; 10:775. [PMID: 31073300 PMCID: PMC6496459 DOI: 10.3389/fimmu.2019.00775] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/25/2019] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) control innate and adaptive immunity by patrolling tissues to gather antigens and danger signals derived from microbes and tissue. Subsequently, DCs integrate those environmental cues, orchestrate immunity or tolerance, and regulate tissue homeostasis. Recent advances in the field of immunometabolism highlight the notion that immune cells markedly alter cellular metabolic pathways during differentiation or upon activation, which has important implications on their functionality. Previous studies showed that active oxidative phosphorylation in mitochondria is associated with immature or tolerogenic DCs, while increased glycolysis upon pathogen sensing can promote immunogenic DC functions. However, new results in the last years suggest that regulation of DC metabolism in steady state, after immunogenic activation and during tolerance in different pathophysiological settings, may be more complex. Moreover, ontogenically distinct DC subsets show different functional specializations to control T cell responses. It is, thus, relevant how metabolism influences DC differentiation and plasticity, and what potential metabolic differences exist among DC subsets. Better understanding of the emerging connection between metabolic adaptions and functional DC specification will likely allow the development of therapeutic strategies to manipulate immune responses.
Collapse
Affiliation(s)
- Stefanie K Wculek
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sofía C Khouili
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Elena Priego
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ignacio Heras-Murillo
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
31
|
Zeng Q, Mallilankaraman K, Schwarz H. Increased Akt-Driven Glycolysis Is the Basis for the Higher Potency of CD137L-DCs. Front Immunol 2019; 10:868. [PMID: 31068941 PMCID: PMC6491642 DOI: 10.3389/fimmu.2019.00868] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 04/04/2019] [Indexed: 12/21/2022] Open
Abstract
CD137 ligand-induced dendritic cells (CD137L-DCs) are a new type of dendritic cells (DCs) that induce strong cytotoxic T cell responses. Investigating the metabolic activity as a potential contributing factor for their potency, we find a significantly higher rate of glycolysis in CD137L-DCs than in granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin 4 induced monocyte-derived DCs (moDCs). Using unbiased screening, Akt-mTORC1 activity was found to be significantly higher throughout the differentiation and maturation of CD137L-DCs than that of moDCs. Furthermore, this higher activity of the Akt-mTORC1 pathway is responsible for the significantly higher glycolysis rate in CD137L-DCs than in moDCs. Inhibition of Akt during maturation or inhibition of glycolysis during and after maturation resulted in suppression of inflammatory DCs, with mature CD137L-DCs being the most affected ones. mTORC1, instead, was indispensable for the differentiation of both CD137L-DCs and moDCs. In contrast to its role in supporting lipid synthesis in murine bone marrow-derived DCs (BMDCs), the higher glycolysis rate in CD137L-DCs does not lead to a higher lipid content but rather to an accumulation of succinate and serine. These data demonstrate that the increased Akt-driven glycolysis underlies the higher activity of CD137L-DCs.
Collapse
Affiliation(s)
- Qun Zeng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Karthik Mallilankaraman
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
32
|
He Z, Zhu X, Shi Z, Wu T, Wu L. Metabolic Regulation of Dendritic Cell Differentiation. Front Immunol 2019; 10:410. [PMID: 30930893 PMCID: PMC6424910 DOI: 10.3389/fimmu.2019.00410] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 02/15/2019] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DCs) are important antigen-presenting cells (APCs) that play essential roles in bridging innate and adaptive immune responses. Differentiation stages of DC subsets from bone marrow progenitor cells have been well-defined during the past decades. Features that distinguish DC progenitor cells from each differentiation stages, related signaling pathways and transcription factors that are crucial for DC lineage commitment have been well-elucidated in numerous studies. Recently, growing evidence are showing that cellular metabolism, as one of the most fundamental process of cells, has essential role in the modulation of immune system. There have been multiple reports and reviews that focus on the metabolic modulations on DC functions, however little attention had been paid to the metabolic regulation of DC development and differentiation. In recent years, increasing evidence suggests that metabolic regulations also exert significant impact on DC differentiation, as well as on the homeostasis of tissue resident DCs. The focus of this review is to summarize the findings from recent studies on the metabolic regulation of DC differentiation and to discuss the impacts of the three major aspects of metabolism on the processes of DC development and differentiation, namely the changes in metabolic pathways, the molecular signaling pathways that modulate cell metabolism, and the effects of metabolites and nutrients. The aim of this review is to draw attentions to this important and exciting research field where the effects of metabolic process and their regulation in DC differentiation need to be further explored.
Collapse
Affiliation(s)
- Zhimin He
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Science, Beijing, China
| | - Xinyi Zhu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
| | - Zhen Shi
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
| | - Tao Wu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
| | - Li Wu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Science, Beijing, China
| |
Collapse
|
33
|
Gilardini Montani MS, Santarelli R, Granato M, Gonnella R, Torrisi MR, Faggioni A, Cirone M. EBV reduces autophagy, intracellular ROS and mitochondria to impair monocyte survival and differentiation. Autophagy 2018; 15:652-667. [PMID: 30324853 DOI: 10.1080/15548627.2018.1536530] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
EBV has been reported to impair monocyte in vitro differentiation into dendritic cells (DCs) and reduce cell survival. In this study, we added another layer of knowledge to this topic and showed that these effects correlated with macroautophagy/autophagy, ROS and mitochondrial biogenesis reduction. Of note, autophagy and ROS, although strongly interconnected, have been separately reported to be induced by CSF2/GM-CSF (colony stimulating factor 2) and required for CSF2-IL4-driven monocyte in vitro differentiation into DCs. We show that EBV infects monocytes and initiates a feedback loop in which, by inhibiting autophagy, reduces ROS and through ROS reduction negatively influences autophagy. Mechanistically, autophagy reduction correlated with the downregulation of RAB7 and ATG5 expression and STAT3 activation, leading to the accumulation of SQSTM1/p62. The latter activated the SQSTM1-KEAP1- NFE2L2 axis and upregulated the anti-oxidant response, reducing ROS and further inhibiting autophagy. ROS decrease correlated also with the reduction of mitochondria, the main source of intracellular ROS, achieved by the downregulation of NRF1 and TFAM, mitochondrial biogenesis transcription factors. Interestingly, mitochondria supply membranes and ATP required for autophagy execution, thus their reduction may further reduce autophagy in EBV-infected monocytes. In conclusion, this study shows for the first time that the interconnected reduction of autophagy, intracellular ROS and mitochondria mediated by EBV switches monocyte differentiation into apoptosis, giving new insights into the mechanisms through which this virus reduces immune surveillance. Abbreviations: ACTB: actin beta; ATG5: autophagy related 5; BAF: bafilomycin A1; BECN1: beclin 1; CAT: catalase; CSF2: colony stimulating factor 2; CT: control; CYCS (cytochrome C: somatic); DCs: dendritic cells; EBV: Epstein-Barr virus; GSR: glutathione-disulfide reductase; KEAP1: kelch like ECH associated protein 1; IL4: interleukin 4; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MET: metformin; NAC: N-acetylcysteine; NFE2L2/NRF2 nuclear factor: erythroid 2 like 2; NRF1 (nuclear respiratory factor 1); clPARP1: cleaved poly(ADP-ribose) polymerase; Rapa: Rapamycin; ROS: reactive oxygen species; SQSTM1/p62: sequestosome 1; TFAM: (transcription factor A: mitochondrial); TUBA1A: tubulin alpha 1a.
Collapse
Affiliation(s)
- M S Gilardini Montani
- a Department of Experimental Medicine , Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti , Rome , Italy
| | - R Santarelli
- a Department of Experimental Medicine , Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti , Rome , Italy
| | - M Granato
- a Department of Experimental Medicine , Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti , Rome , Italy
| | - R Gonnella
- a Department of Experimental Medicine , Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti , Rome , Italy
| | - M R Torrisi
- b Department of Clinical and Molecular Medicine , Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti , Italy.,c Sant'Andrea University Hospital , Azienda Ospedaliera Sant'Andrea , Rome , Italy
| | - A Faggioni
- a Department of Experimental Medicine , Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti , Rome , Italy
| | - M Cirone
- a Department of Experimental Medicine , Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti , Rome , Italy
| |
Collapse
|
34
|
Dominguez-Andres J, Netea MG. Long-term reprogramming of the innate immune system. J Leukoc Biol 2018; 105:329-338. [PMID: 29999546 DOI: 10.1002/jlb.mr0318-104r] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/09/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022] Open
Abstract
During the last few years, a growing body of evidence has shown that immunological memory is not an exclusive trait of lymphocytes, as many inflammatory insults can alter the functionality and the responsiveness of the innate immune system in the long term. Innate immune cells, such as monocytes, macrophages, dendritic cells, and NK cells can be influenced by the encounters with inflammatory stimuli, undergoing functional reprogramming and developing changed responses to subsequent chellenges. The long-term reprogramming depends on the rewiring of cell metabolism and epigenetic processes, and they stay at the basis of induction of both innate immune memory (also termed trained immunity) and innate immune tolerance. Here, we review the central role that the effects of this long-term reprogramming of innate immune cells plays in a number of clinically relevant conditions such as vaccination, atherosclerosis, sepsis, and cancer.
Collapse
Affiliation(s)
- Jorge Dominguez-Andres
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious diseases (RCI), Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.,Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| |
Collapse
|
35
|
Aging impairs mitochondrial respiratory capacity in classical monocytes. Exp Gerontol 2018; 108:112-117. [PMID: 29655929 DOI: 10.1016/j.exger.2018.04.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 12/27/2022]
Abstract
Aging is a critical healthcare concern, with age-related inflammation disposing individuals to a variety of diseases. Monocytes are affected by the aging process, with increased inflammation and impaired cellular functions such as phagocytosis. Mechanisms by which aging alters monocyte function are unknown, but recent research suggests that the balance of metabolic processes determine immune cell phenotype and function. Given the known association between aging and mitochondrial dysfunction in other tissues, we hypothesized that aging would impair mitochondrial function in monocytes. To test this, we isolated classical monocytes from young and older adults and tested mitochondrial function by a Seahorse assay. Aging reduced mitochondrial respiratory capacity and spare capacity in monocytes. Mitochondrial dysfunction is a potential mechanism by which aging alters monocyte phenotype and may impair inflammatory functions, especially in low-glucose environments where oxidative metabolism is necessary to meet energy demands.
Collapse
|
36
|
Dress RJ, Wong AYW, Ginhoux F. Homeostatic control of dendritic cell numbers and differentiation. Immunol Cell Biol 2018; 96:463-476. [DOI: 10.1111/imcb.12028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Regine J Dress
- Singapore Immunology Network (SIgN); Agency for Science; Technology, and Research (A*STAR); Singapore 138648 Singapore
| | - Alicia YW Wong
- Singapore Immunology Network (SIgN); Agency for Science; Technology, and Research (A*STAR); Singapore 138648 Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN); Agency for Science; Technology, and Research (A*STAR); Singapore 138648 Singapore
| |
Collapse
|
37
|
Selleri S, Bifsha P, Civini S, Pacelli C, Dieng MM, Lemieux W, Jin P, Bazin R, Patey N, Marincola FM, Moldovan F, Zaouter C, Trudeau LE, Benabdhalla B, Louis I, Beauséjour C, Stroncek D, Le Deist F, Haddad E. Human mesenchymal stromal cell-secreted lactate induces M2-macrophage differentiation by metabolic reprogramming. Oncotarget 2017; 7:30193-210. [PMID: 27070086 PMCID: PMC5058674 DOI: 10.18632/oncotarget.8623] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 03/26/2016] [Indexed: 12/13/2022] Open
Abstract
Human mesenchymal stromal cells (MSC) have been shown to dampen immune response and promote tissue repair, but the underlying mechanisms are still under investigation. Herein, we demonstrate that umbilical cord-derived MSC (UC-MSC) alter the phenotype and function of monocyte-derived dendritic cells (DC) through lactate-mediated metabolic reprogramming. UC-MSC can secrete large quantities of lactate and, when present during monocyte-to-DC differentiation, induce instead the acquisition of M2-macrophage features in terms of morphology, surface markers, migratory properties and antigen presentation capacity. Microarray expression profiling indicates that UC-MSC modify the expression of metabolic-related genes and induce a M2-macrophage expression signature. Importantly, monocyte-derived DC obtained in presence of UC-MSC, polarize naïve allogeneic CD4+ T-cells into Th2 cells. Treatment of UC-MSC with an inhibitor of lactate dehydrogenase strongly decreases lactate concentration in culture supernatant and abrogates the effect on monocyte-to-DC differentiation. Metabolic analysis further revealed that UC-MSC decrease oxidative phosphorylation in differentiating monocytes while strongly increasing the spare respiratory capacity proportional to the amount of secreted lactate. Because both MSC and monocytes are recruited in vivo at the site of tissue damage and inflammation, we propose the local increase of lactate concentration induced by UC-MSC and the consequent enrichment in M2-macrophage generation as a mechanism to achieve immunomodulation.
Collapse
Affiliation(s)
- Silvia Selleri
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, QC, Canada
| | - Panojot Bifsha
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, QC, Canada
| | - Sara Civini
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Consiglia Pacelli
- Department of Pharmacology, University of Montreal, Montreal, QC, Canada
| | - Mame Massar Dieng
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Biology, New York University, Abu Dhabi, United Arab Emirates
| | - William Lemieux
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, QC, Canada
| | - Ping Jin
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Renée Bazin
- Department of Research and Development, Héma-Québec, Québec, QC, Canada
| | - Natacha Patey
- Department of Pathology, University of Montreal, Montreal, QC, Canada
| | - Francesco M Marincola
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA.,Sidra Medical and Research Center, Doha, Qatar
| | - Florina Moldovan
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Faculty of Dentistry, University of Montreal, Montreal, QC, Canada
| | | | - Louis-Eric Trudeau
- Department of Pharmacology, University of Montreal, Montreal, QC, Canada
| | | | - Isabelle Louis
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Christian Beauséjour
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Pharmacology, University of Montreal, Montreal, QC, Canada
| | - David Stroncek
- Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Françoise Le Deist
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, QC, Canada.,Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Elie Haddad
- CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, QC, Canada.,Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
38
|
Metabolic Plasticity in Dendritic Cell Responses: Implications in Allergic Asthma. J Immunol Res 2017; 2017:5134760. [PMID: 29387732 PMCID: PMC5745769 DOI: 10.1155/2017/5134760] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/07/2017] [Indexed: 12/25/2022] Open
Abstract
Dendritic cells (DCs) are highly specialized in antigen presentation and play a pivotal role in the initiation, progression, and perpetuation of adaptive immune responses. Emerging immune pathways are being recognized increasingly for DCs and their subsets that differentially regulate T lymphocyte function based on the type and interactions with the antigen. However, these interactions not only alter the signaling process and DC function but also render metabolic plasticity. The current review focuses on the metabolic cues of DCs that coordinate DC activation and differentiation and discuss whether targeting these fundamental cellular processes have implications to control airway inflammation and adaptive immunity. Therefore, strategies using metabolism-based therapeutic manipulation of DC functions could be developed into novel treatments for airway inflammation and asthma.
Collapse
|
39
|
Singh A, Sen E. Reciprocal role of SIRT6 and Hexokinase 2 in the regulation of autophagy driven monocyte differentiation. Exp Cell Res 2017; 360:365-374. [PMID: 28935467 DOI: 10.1016/j.yexcr.2017.09.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/16/2022]
Abstract
Emerging evidences suggest the impact of autophagy on differentiation but the underlying molecular links between metabolic restructuring and autophagy during monocyte differentiation remain elusive. An increase in PPARγ, HK2 and SIRT6 expression was observed upon PMA induced monocyte differentiation. While PPARγ positively regulated HK2 and SIRT6 expression, the latter served as a negative regulator of HK2. Changes in expression of these metabolic modelers were accompanied by decreased glucose uptake and increase in Chibby, a potent antagonist of β-catenin/Wnt pathway. Knockdown of Chibby abrogated PMA induced differentiation. While inhibition of HK2 either by Lonidamine or siRNA further elevated PMA induced Chibby, mitochondrial ROS, TIGAR and LC3II levels; siRNA mediated knock-down of SIRT6 exhibited contradictory effects as compared to HK2. Notably, inhibition of autophagy increased HK2, diminished Chibby level and CD33 expression. In addition, PMA induced expression of cytoskeletal architectural proteins, CXCR4, phagocytosis, acquisition of macrophage phenotypes and release of pro-inflammatory mediators was found to be HK2 dependent. Collectively, our findings highlight the previously unknown reciprocal influence of SIRT6 and HK2 in regulating autophagy driven monocyte differentiation.
Collapse
Affiliation(s)
- Ankita Singh
- National Brain Research Centre, Manesar, Haryana 122051, India
| | - Ellora Sen
- National Brain Research Centre, Manesar, Haryana 122051, India.
| |
Collapse
|
40
|
Vanherwegen AS, Gysemans C, Mathieu C. Vitamin D endocrinology on the cross-road between immunity and metabolism. Mol Cell Endocrinol 2017; 453:52-67. [PMID: 28461074 DOI: 10.1016/j.mce.2017.04.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/13/2022]
Abstract
The effects of vitamin D on the immune function have been recognized for more than a quarter of a century. However, our understanding of the multifactorial nature of the effects of vitamin D at the cellular, molecular and metabolic level in different immune cells of the innate and adaptive immune system has dramatically progressed during the last decades. In this review, we summarize the main metabolic pathways preferentially used in different subsets of macrophages, dendritic cells, T and B cells as well as the immunomodulatory effects of vitamin D on these cells. We will highlight the metabolic reprogramming happening in vitamin D-conditioned tolerogenic dendritic cells. A better knowledge of the dynamics of metabolic states in immune subsets and their possible roles in inflammation and autoimmunity may advance the development of novel immunotherapies. Likewise, the implications of effects of vitamin D on immunometabolism may progress our insights in the nature of immune responses in health and disease.
Collapse
Affiliation(s)
- An-Sofie Vanherwegen
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Herestraat 49, O&N1, Bus 902, 3000 Leuven, Belgium
| | - Conny Gysemans
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Herestraat 49, O&N1, Bus 902, 3000 Leuven, Belgium.
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Herestraat 49, O&N1, Bus 902, 3000 Leuven, Belgium
| |
Collapse
|
41
|
Schneider KM, Watson NB, Minchenberg SB, Massa PT. The influence of macrophage growth factors on Theiler's Murine Encephalomyelitis Virus (TMEV) infection and activation of macrophages. Cytokine 2017; 102:83-93. [PMID: 28800924 DOI: 10.1016/j.cyto.2017.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023]
Abstract
Macrophages are common targets for infection and innate immune activation by many pathogenic viruses including the neurotropic Theiler's Murine Encephalomyelitis Virus (TMEV). As both infection and innate activation of macrophages are key determinants of viral pathogenesis especially in the central nervous system (CNS), an analysis of macrophage growth factors on these events was performed. C3H mouse bone-marrow cells were differentiated in culture using either recombinant macrophage colony stimulating factor (M-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF), inoculated with TMEV (BeAn) and analyzed at various times thereafter. Cytokine RNA and protein analysis, virus titers, and flow cytometry were performed to characterize virological parameters under these culture conditions. GM-CSF-differentiated macrophages showed higher levels of TMEV viral RNA and proinflammatory molecules compared to infected M-CSF-differentiated cells. Thus, GM-CSF increases both TMEV infection and TMEV-induced activation of macrophages compared to that seen with M-CSF. Moreover, while infectious viral particles decreased from a peak at 12h to undetectable levels at 48h post infection, TMEV viral RNA remained higher in GM-CSF- compared to M-CSF-differentiated macrophages in concert with increased proinflammatory gene expression. Analysis of a possible basis for these differences determined that glycolytic rates contributed to heightened virus replication and proinflammatory cytokine secretion in GM-CSF compared to M-CSF-differentiated macrophages. In conclusion, we provide evidence implicating a role for GM-CSF in promoting virus replication and proinflammatory cytokine expression in macrophages, indicating that GM-CSF may be a key factor for TMEV infection and the induction of chronic TMEV-induced immunopathogenesis in the CNS.
Collapse
Affiliation(s)
- Karin M Schneider
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Neva B Watson
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Scott B Minchenberg
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Paul T Massa
- Department of Neurology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
42
|
|
43
|
Cameron AM, Lawless SJ, Pearce EJ. Metabolism and acetylation in innate immune cell function and fate. Semin Immunol 2016; 28:408-416. [PMID: 28340958 PMCID: PMC10911065 DOI: 10.1016/j.smim.2016.10.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/10/2016] [Accepted: 10/12/2016] [Indexed: 11/29/2022]
Abstract
Innate immunity is the first line of defense against invading pathogens. Changes in both metabolism and chromatin accessibility contribute to the shaping of these innate immune responses, and we are beginning to appreciate that cross-talk between these two systems plays an important role in determining innate immune cell differentiation and function. In this review we focus on acetylation, a post-translational modification important for both regulating chromatin accessibility by modulating histone function, and for functional regulation of non-histone proteins, which has many links to both immune signaling and metabolism. We discuss the interactions between metabolism and acetylation, including the requirement for metabolic intermediates as substrates and co-factors for acetylation, and the regulation of metabolic proteins and enzymes by acetylation. Here we highlight recent findings, which demonstrate the role that the metabolism-acetylation axis has in coordinating the responses of innate immune cells to the availability of nutrients and the microenvironment.
Collapse
Affiliation(s)
- Alanna M Cameron
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Simon J Lawless
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Edward J Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
44
|
Iacovelli J, Rowe GC, Khadka A, Diaz-Aguilar D, Spencer C, Arany Z, Saint-Geniez M. PGC-1α Induces Human RPE Oxidative Metabolism and Antioxidant Capacity. Invest Ophthalmol Vis Sci 2016; 57:1038-51. [PMID: 26962700 PMCID: PMC4788093 DOI: 10.1167/iovs.15-17758] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Oxidative stress and metabolic dysregulation of the RPE have been implicated in AMD; however, the molecular regulation of RPE metabolism remains unclear. The transcriptional coactivator, peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α) is a powerful mediator of mitochondrial function. This study examines the ability of PGC-1α to regulate RPE metabolic program and oxidative stress response. Methods Primary human fetal RPE (hfRPE) and ARPE-19 were matured in vitro using standard culture conditions. Mitochondrial mass of RPE was measured using MitoTracker staining and citrate synthase activity. Expression of PGC-1 isoforms, RPE-specific genes, oxidative metabolism proteins, and antioxidant enzymes was analyzed by quantitative PCR and Western blot. Mitochondrial respiration and fatty-acid oxidation were monitored using the Seahorse extracellular flux analyzer. Expression of PGC-1α was increased using adenoviral delivery. ARPE-19 were exposed to hydrogen peroxide to induce oxidative stress. Reactive oxygen species were measured by CM-H2DCFDA fluorescence. Cell death was analyzed by LDH release. Results Maturation of ARPE-19 and hfRPE was associated with significant increase in mitochondrial mass, expression of oxidative phosphorylation (OXPHOS) genes, and PGC-1α gene expression. Overexpression of PGC-1α increased expression of OXPHOS and fatty-acid β-oxidation genes, ultimately leading to the potent induction of mitochondrial respiration and fatty-acid oxidation. PGC-1α gain of function also strongly induced numerous antioxidant genes and, importantly, protected RPE from oxidant-mediated cell death without altering RPE functions. Conclusions This study provides important insights into the metabolic changes associated with RPE functional maturation and identifies PGC-1α as a potent driver of RPE mitochondrial function and antioxidant capacity.
Collapse
Affiliation(s)
- Jared Iacovelli
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States 2Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Glenn C Rowe
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Arogya Khadka
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States
| | - Daniel Diaz-Aguilar
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States
| | - Carrie Spencer
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States
| | - Zoltan Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Magali Saint-Geniez
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States 2Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
45
|
Romero-Garcia S, Moreno-Altamirano MMB, Prado-Garcia H, Sánchez-García FJ. Lactate Contribution to the Tumor Microenvironment: Mechanisms, Effects on Immune Cells and Therapeutic Relevance. Front Immunol 2016; 7:52. [PMID: 26909082 PMCID: PMC4754406 DOI: 10.3389/fimmu.2016.00052] [Citation(s) in RCA: 343] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/02/2016] [Indexed: 01/12/2023] Open
Abstract
Malignant transformation of cells leads to enhanced glucose uptake and the conversion of a larger fraction of pyruvate into lactate, even under normoxic conditions; this phenomenon of aerobic glycolysis is largely known as the Warburg effect. This metabolic reprograming serves to generate biosynthetic precursors, thus facilitating the survival of rapidly proliferating malignant cells. Extracellular lactate directs the metabolic reprograming of tumor cells, thereby serving as an additional selective pressure. Besides tumor cells, stromal cells are another source of lactate production in the tumor microenvironment, whose role in both tumor growth and the antitumor immune response is the subject of intense research. In this review, we provide an integral perspective of the relationship between lactate and the overall tumor microenvironment, from lactate structure to metabolic pathways for its synthesis, receptors, signaling pathways, lactate-producing cells, lactate-responding cells, and how all contribute to the tumor outcome. We discuss the role of lactate as an immunosuppressor molecule that contributes to tumor evasion and we explore the possibility of targeting lactate metabolism for cancer treatment, as well as of using lactate as a prognostic biomarker.
Collapse
Affiliation(s)
- Susana Romero-Garcia
- Chronic-Degenerative Department, National Institute of Respiratory Diseases "Ismael Cosio Villegas" , Mexico City , Mexico
| | - María Maximina B Moreno-Altamirano
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional , Mexico City , Mexico
| | - Heriberto Prado-Garcia
- Chronic-Degenerative Department, National Institute of Respiratory Diseases "Ismael Cosio Villegas" , Mexico City , Mexico
| | - Francisco Javier Sánchez-García
- Laboratorio de Inmunorregulación, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional , Mexico City , Mexico
| |
Collapse
|
46
|
Metabolism Is Central to Tolerogenic Dendritic Cell Function. Mediators Inflamm 2016; 2016:2636701. [PMID: 26980944 PMCID: PMC4766347 DOI: 10.1155/2016/2636701] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 12/31/2015] [Indexed: 12/13/2022] Open
Abstract
Immunological tolerance is a fundamental tenant of immune homeostasis and overall health. Self-tolerance is a critical component of the immune system that allows for the recognition of self, resulting in hyporeactivity instead of immunogenicity. Dendritic cells are central to the establishment of dominant immune tolerance through the secretion of immunosuppressive cytokines and regulatory polarization of T cells. Cellular metabolism holds the key to determining DC immunogenic or tolerogenic cell fate. Recent studies have demonstrated that dendritic cell maturation leads to a shift toward a glycolytic metabolic state and preferred use of glucose as a carbon source. In contrast, tolerogenic dendritic cells favor oxidative phosphorylation and fatty acid oxidation. This dichotomous metabolic reprogramming of dendritic cells drives differential cellular function and plays a role in pathologies, such as autoimmune disease. Pharmacological alterations in metabolism have promising therapeutic potential.
Collapse
|
47
|
Aquilano K, Lettieri Barbato D, Rosa CM. The multifaceted role of nitric oxide synthases in mitochondrial biogenesis and cell differentiation. Commun Integr Biol 2015; 8:e1017158. [PMID: 26479127 PMCID: PMC4594549 DOI: 10.1080/19420889.2015.1017158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/04/2014] [Indexed: 10/25/2022] Open
Abstract
Nitric oxide (NO) is physiologically synthetized by a family of enzymes called NO synthases (NOSs). NO is a pleiotropic second messenger having a fundamental role in several cellular processes including cell differentiation. Being a high reactive molecule, NO must be synthetized in close proximity to the effector/target. For this reason, the subcellular localization of NOSs is tightly regulated by different post-translation mechanisms. Recently, in murine C2C12 myoblasts, we have demonstrated that mitochondrial biogenesis, an essential event for cell differentiation, can be effective only if the site of NO production is located at nuclear level, where NO favors the CREB-dependent expression of PGC-1α gene. The increase of NO flux in nuclei is elicited by the up-regulation and redistribution of neuronal NOS (nNOS) toward nuclei. Herein we show that an upregulation of endothelial NOS (eNOS) occurs during adipocyte differentiation in 3T3-L1 cells. However, differently to differentiating myocytes, a concomitant redistribution of eNOS toward nuclei was not detected. We also observed that, upon treatment with the NO synthesis inhibitor L-NAME, mitochondrial biogenesis as well as triglyceride accumulation that normally occurs during adipogenesis were not impeded. The absence of eNOS in nuclei together with the ineffectiveness of L-NAME suggest that, at least during 3T3-L1 differentiation, NO is not fundamental for the induction of mitochondrial biogenesis and adipogenesis.
Collapse
Affiliation(s)
- Katia Aquilano
- Department of Biology; University of Rome "Tor Vergata" ; Rome, Italy ; IRCCS San Raffaele ; Rome, Italy
| | | | - Ciriolo Maria Rosa
- Department of Biology; University of Rome "Tor Vergata" ; Rome, Italy ; IRCCS San Raffaele ; Rome, Italy
| |
Collapse
|
48
|
Abstract
The past 15 years have seen enormous advances in our understanding of the receptor and signalling systems that allow dendritic cells (DCs) to respond to pathogens or other danger signals and initiate innate and adaptive immune responses. We are now beginning to appreciate that many of these pathways not only stimulate changes in the expression of genes that control DC immune functions, but also affect metabolic pathways, thereby integrating the cellular requirements of the activation process. In this Review, we focus on this relatively new area of research and attempt to describe an integrated view of DC immunometabolism.
Collapse
Affiliation(s)
- Edward J Pearce
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
49
|
Ryu SW, Han EC, Yoon J, Choi C. The mitochondrial fusion-related proteins Mfn2 and OPA1 are transcriptionally induced during differentiation of bone marrow progenitors to immature dendritic cells. Mol Cells 2015; 38:89-94. [PMID: 25387754 PMCID: PMC4314123 DOI: 10.14348/molcells.2015.2285] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/04/2014] [Indexed: 02/04/2023] Open
Abstract
The shape and activity of mitochondria are tightly regulated by fusion and fission processes that are essential for maintaining normal cellular function. However, little is known about the involvement of mitochondrial dynamics in the development of the immune system. In this study, we demonstrate that mitochondrial dynamics play a role in the differentiation and migration of immature dendritic cells (imDCs). We show that mitochondrial elongation is induced during GM-CSF-stimulated differentiation of bone marrow progenitors to imDCs accompanied by upregulation of mitochondrial fusion proteins. These processes precede the changes in mitochondrial morphology and connectivity that occur during differentiation. Mfn2 and OPA1, but not Mfn1, are transcriptionally upregulated during differentiation; however, knockdown of Mfn2 and OPA1 does not induce any change in expression of CD11c, CDC80, or CD86. Notably, knockdown of Mfn2 or OPA1 by siRNA in imDCs significantly reduces CCR7 expression and CCL19-mediated migration. These results suggest that the mitochondrial fusion-related proteins Mfn2 and OPA1 are upregulated during bone marrow progenitor differentiation and promote the migration of imDCs by regulating the expression of CCR7.
Collapse
Affiliation(s)
- Seung-Wook Ryu
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
- KI for the Biocentury, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Eun Chun Han
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Jonghee Yoon
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Chulhee Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
- KI for the Biocentury, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| |
Collapse
|
50
|
Luo C, Shen G, Liu N, Gong F, Wei X, Yao S, Liu D, Teng X, Ye N, Zhang N, Zhou X, Li J, Yang L, Zhao X, Yang L, Xiang R, Wei YQ. Ammonia Drives Dendritic Cells into Dysfunction. THE JOURNAL OF IMMUNOLOGY 2014; 193:1080-9. [DOI: 10.4049/jimmunol.1303218] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|