1
|
Amiri SP, Nejad FS, Karamigolbaghi M, Jafari E, Robat-Jazi B, Sadeghi A, Amiri SGP, Namdari H, Saboor-Yaraghi AA. Decreased circulating CD39+ regulatory T cell frequencies following non-traumatic brain death. Transpl Immunol 2025; 90:102219. [PMID: 40118258 DOI: 10.1016/j.trim.2025.102219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND AND PURPOSE Regulatory T cells (Tregs) are thought to modulate immune responses during Brain death (BD), However findings on their role remain controversial. This study aimed to assess the frequency of circulating Tregs in the peripheral blood of non-traumatic BD cases, specifically focusing on CD4+CD25+CD127low/-CD39+ Tregs and the levels of inflammatory cytokine mRNA in BD individuals. METHODS The percentage of CD4+CD25+CD127low/-CD39+ Tregs was measured using flow cytometry in BD patients upon admission and in control subjects. Additionally, mRNA expression levels of interleukin (IL)-1β, IL-6, IL-8, IL-17, tumor necrosis factor (TNF)-α and Interferon (IFN)-γ were quantified in peripheral blood mononuclear cells (PBMCs) from 28 BD individuals and 28 controls using real-time polymerase chain reaction. RESULTS CD39+ Tregs were significantly reduced in non-traumatic BD cases compared with control group (P < 0.0001). Moreover, the expression levels of IL-1β, IL-6, IL-8, IL-17a, IFN-ɣ, and TNF-α were significantly elevated in non-traumatic BD cases compared to the control group (P < 0.01, P < 0.05, P < 0.01, P < 0.0001, P < 0.0001, P < 0.001 respectively). CONCLUSION This study provides novel evidence of reduced CD39+ Tregs in the peripheral blood of non-traumatic BD patients, accompanied by increased inflammatory cytokine gene expression. Further investigations are needed to explore the underlying mechanisms and potential therapeutic implications.
Collapse
Affiliation(s)
- Sedighe Poursaleh Amiri
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran; Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fattah Sotoudeh Nejad
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Karamigolbaghi
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Jafari
- Department of Stem Cells Technology and Tissue Regeneration, School of Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Robat-Jazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Iran
| | - Ahmadreza Sadeghi
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ghasem Poursaleh Amiri
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Haideh Namdari
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
2
|
Rabbani P, Ramkhelawon B, Cronstein BN. Adenosine metabolism and receptors in aging of the skin, musculoskeletal, immune and cardiovascular systems. Ageing Res Rev 2025; 106:102695. [PMID: 39971100 PMCID: PMC11960428 DOI: 10.1016/j.arr.2025.102695] [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: 07/22/2024] [Revised: 11/26/2024] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Aging populations worldwide face an increasing burden of age-related chronic conditions, necessitating a deeper understanding of the underlying mechanisms. Purine metabolism has emerged as a crucial player in the pathophysiology of aging, affecting various tissues and organs. Dysregulation of purine metabolism, particularly alterations in extracellular adenosine levels and adenosine receptor signaling, contributes to age-related musculoskeletal problems, cardiovascular diseases, inflammation, and impaired immune responses. Changes in purine metabolism are associated with diminished tissue repair and regeneration, altered bone density, and impaired muscle regeneration. Mechanistically, age-related alterations in purine metabolism involve reductions in extracellular adenosine production, impaired autocrine signaling, and dysregulated expression of CD73 and CD39. Targeting adenosine receptors, such as A2A and A2B receptors, emerges as a promising therapeutic approach to mitigate age-related conditions, including sarcopenia, obesity, osteoarthritis, and impaired wound healing. Since we cannot reverse time, understanding the intricate molecular interplay between purine metabolism and aging-related pathologies holds significant potential for developing novel therapeutic strategies to improve the health and quality of life of aging populations. In this review, we compile the findings related to purine metabolism during aging in several tissues and organs and provide insights into how these signals can be manipulated to circumvent the deleterious effects of the passage of time on our body.
Collapse
Affiliation(s)
- Piul Rabbani
- Hansjorg Wyss Department of Plastic Surgery, New York University Langone Health, New York, NY, USA
| | - Bhama Ramkhelawon
- Division of Vascular Surgery, Department of Surgery, New York University Langone Health, New York, NY, USA; Department of Cell Biology, New York University Langone Health, New York, NY, USA
| | - Bruce N Cronstein
- Department of Medicine, Divisions of Rheumatology and Precision Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
3
|
Liu R, Tang L, Liu Y, Hu H, Liu J. Causal relationship between immune cell signatures and colorectal cancer: a bi-directional, two-sample mendelian randomization study. BMC Cancer 2025; 25:387. [PMID: 40033246 PMCID: PMC11877943 DOI: 10.1186/s12885-025-13576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Prior studies have demonstrated the association between immune cells and colorectal cancer (CRC). However, the causal link to specific immunophenotypes is limited. This study intends to elucidate the causal relationship of immune cell signatures on CRC. METHODS We performed a bi-directional and two-sample mendelian randomization (MR) study, utilizing GWAS summary data of 731 immune cell traits (n = 3,757) and CRC statistics (n = 470,002). The primary MR methodology was inverse-variance weighted (IVW) method. Furthermore, heterogeneity was evaluated by Cochran's Q test. MR-PRESSO and MR-Egger were employed to assess horizontal and vertical pleiotropy respectively. Sensitivity analysis and FDR correction were conducted in our results. These results were validated in both the UK Biobank and FinnGen cohorts. We also extracted transcriptomic data of CRC and adjacent non-tumor tissues from TCGA, and used CIBERSORT to compare the infiltration patterns of 22 immune cell panels between normal tissues and the tumor microenvironment (TME). RESULTS Our study indicated nine immune cell signatures had significant causality with the risk of CRC after sensitivity analysis and FDR correction. The positive results covered four panels: B cell, CD8 + T cell, Treg, and monocyte. IgD- CD38br and IgD + CD38br B cell, CD8dim and CD28 + CD45RA- CD8dim T cell, and CD14 on CD14 + CD16- monocyte were the protective factors of CRC. However, CD39 + resting Treg, CX3CR1 on CD14- CD16 + monocyte, FSC-A on HLA DR + T cell, and BAFF-R on B cell increased the risk of CRC. The results were validated in the UK Biobank data and FinnGen cohorts. The data from the TCGA database also confirmed the infiltration of B cell, CD8 + T cell, Treg, and monocyte panels in the TME. CONCLUSION This study highlights the causal link between specific immune cell phenotypes and CRC, providing valuable insights into the immune microenvironment's role in CRC. The validation of our findings using large-scale datasets (UK Biobank, FinnGen) and TCGA underscores the robustness of our results, offering new potential therapeutic targets for CRC treatment.
Collapse
Affiliation(s)
- Ruizhi Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liansha Tang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, Sichuan Province, 610041, China
| | - Yunjia Liu
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Handan Hu
- Queen Mary College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, 37 Guoxue Xiang Street, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
4
|
Toller-Kawahisa JE, Viacava PR, Palsson-McDermott EM, Nascimento DC, Cervantes-Silva MP, O'Carroll SM, Zotta A, Damasceno LEA, Públio GA, Forti P, Luiz JPM, Silva de Melo BM, Martins TV, Faça VM, Curtis A, Cunha TM, Cunha FDQ, O'Neill LAJ, Alves-Filho JC. Metabolic reprogramming of macrophages by PKM2 promotes IL-10 production via adenosine. Cell Rep 2025; 44:115172. [PMID: 39772395 PMCID: PMC11781862 DOI: 10.1016/j.celrep.2024.115172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/24/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Macrophages play a crucial role in immune responses and undergo metabolic reprogramming to fulfill their functions. The tetramerization of the glycolytic enzyme pyruvate kinase M2 (PKM2) induces the production of the anti-inflammatory cytokine interleukin (IL)-10 in vivo, but the underlying mechanism remains elusive. Here, we report that PKM2 activation with the pharmacological agent TEPP-46 increases IL-10 production in LPS-activated macrophages by metabolic reprogramming, leading to the production and release of ATP from glycolysis. The effect of TEPP-46 is abolished in PKM2-deficient macrophages. Extracellular ATP is converted into adenosine by ectonucleotidases that activate adenosine receptor A2a (A2aR) to enhance IL-10 production. Interestingly, IL-10 production induced by PKM2 activation is associated with improved mitochondrial health. Our results identify adenosine derived from glycolytic ATP as a driver of IL-10 production, highlighting the role of tetrameric PKM2 in regulating glycolysis to promote IL-10 production.
Collapse
Affiliation(s)
- Juliana Escher Toller-Kawahisa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Paula Ramos Viacava
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Daniele Carvalho Nascimento
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Mariana Patricia Cervantes-Silva
- School of Pharmacy and Biomolecular Sciences and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Shane Myles O'Carroll
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Alessia Zotta
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Luis Eduardo Alves Damasceno
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Gabriel Azevedo Públio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Pedro Forti
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - João Paulo Mesquita Luiz
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Bruno Marcel Silva de Melo
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Timna Varela Martins
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Annie Curtis
- School of Pharmacy and Biomolecular Sciences and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fernando de Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Luke Anthony John O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland.
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
5
|
Gockeln L, Wirsdörfer F, Jendrossek V. CD73/adenosine dynamics in treatment-induced pneumonitis: balancing efficacy with risks of adverse events in combined radio-immunotherapies. Front Cell Dev Biol 2025; 12:1471072. [PMID: 39872847 PMCID: PMC11769960 DOI: 10.3389/fcell.2024.1471072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Consolidation with PD-1/PD-L1-based immune checkpoint blockade after concurrent platinum-based chemo-radiotherapy has become the new standard of care for advanced stage III unresectable non-small cell lung cancer (NSCLC) patients. In order to further improve therapy outcomes, innovative combinatorial treatment strategies aim to target additional immunosuppressive barriers in the tumor microenvironment such as the CD73/adenosine pathway. CD73 and adenosine are known as crucial endogenous regulators of lung homeostasis and inflammation, but also contribute to an immunosuppressive tumor microenvironment. Furthermore, the CD73/adenosine pathway can also limit the immune-activating effects of cytotoxic therapies by degrading the pro-inflammatory danger molecule ATP, which is released into the tumor microenvironment and normal lung tissue upon therapy-induced cell damage. Thus, while targeting CD73 may enhance the efficacy of radio-immunotherapies in cancer treatment by mitigating tumor immune escape and improving immune-mediated tumor killing, it also raises concerns about increased immune-related adverse events (irAEs) in the normal tissue. In fact, combined radio-immunotherapies bear an increased risk of irAEs in the lungs, and additional pharmacologic inhibition of CD73 may further enhance the risk of overwhelming or overlapping pulmonary toxicity and thereby limit therapy outcome. This review explores how therapeutic interventions targeting CD73/adenosine dynamics could enhance radiation-induced immune activation in combined radio-immunotherapies, whilst potentially driving irAEs in the lung. We specifically investigate the interactions between radiotherapy and the CD73/adenosine pathway in radiation pneumonitis. Additionally, we compare the incidence of (radiation) pneumonitis reported in relevant trials to determine if there is an increased risk of irAEs in the clinical setting. By understanding these dynamics, we aim to inform future strategies for optimizing radio-immunotherapy regimens, ensuring effective cancer control while preserving pulmonary integrity and patient quality of life.
Collapse
Affiliation(s)
| | | | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
6
|
Li X, Zhou B, Xu F, Liu H, Jia X. Causal effect of immune cells on idiopathic pulmonary fibrosis: A mendelian randomization study. Heart Lung 2024; 68:9-17. [PMID: 38865855 DOI: 10.1016/j.hrtlng.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND A key component of idiopathic pulmonary fibrosis (IPF) is the involvement of immune cells. However, the causal interaction between different immune cell signatures and IPF remain inconclusive. OBJECTIVES Based on publicly accessible data, our study utilized the Mendelian randomization (MR) approach to determine the causative relevance of complex immune cell phenotypes in IPF. METHODS We deployed a two-sample Mendelian randomization approach to evaluate the causal interaction between immune cell markers and IPF. All data regarding 731 immune signatures and IPF were acquired from two genome-wide association studies (GWAS) that are accessible to the public. The original study adopted the inverse variance weighted (IVW) method, followed by sensitivity analyses aimed at eliminating heterogeneity and pleiotropy. Additionally, Multivariate Mendelian randomization (MVMR) was utilized to identify the independent risk factors in our study. RESULTS The summary dataset for IPF was accessed from the Finnish Genetic Consortium R9, comprising 2018 patients and 373,064 controls. And the dataset for immune signatures was conducted in 3,757 Sardinian individuals. By conducting IVW and extensive sensitivity analyses, univariate Mendelian randomization (UVMR) identified one immunophenotype that remained causally associated with IPF after false discovery rate (FDR) correction: CD39 on CD39+ CD8+T cells (odd ratio [OR] = 0.850, 95 % confidence interval [CI] = 0.787-0.918, P = 3.68 × 10-5). The causal association with IPF was further validated using MVMR. CONCLUSIONS Based on rigorous MR analysis methods and FDR correction, our study demonstrated that CD39 on CD39+ CD8+T cells showed a protective effect against IPF, providing effective insights for preventing and diagnosing pulmonary fibrosis.
Collapse
Affiliation(s)
- Xuannian Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Bowen Zhou
- The First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Fei Xu
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huaman Liu
- Department of General Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinhua Jia
- Department of Pneumology and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
7
|
Hegazi A, Rager LE, Watkins DE, Su KH. Advancing Immunotherapy in Pancreatic Cancer. Int J Mol Sci 2024; 25:11560. [PMID: 39519112 PMCID: PMC11546161 DOI: 10.3390/ijms252111560] [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: 10/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic cancer remains one of the deadliest malignancies, with a consistently low five-year survival rate for the past several decades. This is in stark contrast to other cancers, which have seen significant improvement in survival and prognosis due to recent developments in therapeutic modalities. These modest improvements in pancreatic cancer outcomes have primarily resulted from minor advances in cytotoxic chemotherapeutics, with limited progress in other treatment approaches. A major focus of current therapeutic research is the further development of immunomodulatory therapies characterized by antibody-based approaches, cellular therapies, and vaccines. Although initial results utilizing immunotherapy in pancreatic cancer have been mixed, recent clinical trials have demonstrated significant improvements in patient outcomes. In this review, we detail these three approaches to immunomodulation, highlighting their common targets and distinct shortcomings, and we provide a narrative summary of completed and ongoing clinical trials that utilize these approaches to immunomodulation. Within this context, we aim to inform future research efforts by identifying promising areas that warrant further exploration.
Collapse
Affiliation(s)
| | | | | | - Kuo-Hui Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (A.H.); (L.E.R.); (D.E.W.)
| |
Collapse
|
8
|
Laketa D, Lavrnja I. Extracellular Purine Metabolism-Potential Target in Multiple Sclerosis. Mol Neurobiol 2024; 61:8361-8386. [PMID: 38499905 DOI: 10.1007/s12035-024-04104-9] [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: 07/26/2023] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
The purinergic signaling system comprises a complex network of extracellular purines and purine-metabolizing ectoenzymes, nucleotide and nucleoside receptors, ATP release channels, and nucleoside transporters. Because of its immunomodulatory function, this system is critically involved in the pathogenesis of multiple sclerosis (MS) and its best-characterized animal model, experimental autoimmune encephalomyelitis (EAE). MS is a chronic neuroinflammatory demyelinating and neurodegenerative disease with autoimmune etiology and great heterogeneity, mostly affecting young adults and leading to permanent disability. In MS/EAE, alterations were detected in almost all components of the purinergic signaling system in both peripheral immune cells and central nervous system (CNS) glial cells, which play an important role in the pathogenesis of the disease. A decrease in extracellular ATP levels and an increase in its downstream metabolites, particularly adenosine and inosine, were frequently observed at MS, indicating a shift in metabolism toward an anti-inflammatory environment. Accordingly, upregulation of the major ectonucleotidase tandem CD39/CD73 was detected in the blood cells and CNS of relapsing-remitting MS patients. Based on the postulated role of A2A receptors in the transition from acute to chronic neuroinflammation, the association of variants of the adenosine deaminase gene with the severity of MS, and the beneficial effects of inosine treatment in EAE, the adenosinergic system emerged as a promising target in neuroinflammation. More recently, several publications have identified ADP-dependent P2Y12 receptors and the major extracellular ADP producing enzyme nucleoside triphosphate diphosphohydrolase 2 (NTPDase2) as novel potential targets in MS.
Collapse
Affiliation(s)
- Danijela Laketa
- Department of General Physiology and Biophysics, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Studentski Trg 3, Belgrade, Republic of Serbia.
| | - Irena Lavrnja
- Institute for Biological Research, Sinisa Stankovic" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade, Republic of Serbia
| |
Collapse
|
9
|
Doleski PH, Cabral FL, Jantsch MH, Ebone RS, Adefegha SA, Leal DBR, Schetinger MRC. Kinetic properties of E-NTPDase activity in lymphocytes isolated from bone marrow, thymus and mesenteric lymph nodes of Wistar rats. Mol Cell Biochem 2024; 479:2447-2458. [PMID: 37792238 DOI: 10.1007/s11010-023-04860-7] [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: 07/24/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023]
Abstract
Plasma membrane anchored nucleotidases (E-ATPDases), as the E-NTPDase family, could hydrolyze and regulate the pericellular levels of nucleotides in lymphocytes. Each immune organ has a different microenvironment and display lymphocytes with different functions and phenotypes. Considering the different functions of each resident subpopulations of lymphocytes, the E-ATPDases activities in bone marrow (BML), thymus (TL) and mesenteric lymph node (MLL) lymphocytes of Wistar rats were characterized. The hydrolysis of extracellular nucleotides (eATP and eADP) showed linearity in time of reaction between 0 and 120 min, and concentration of lymphocytes expressed in proteins between 1 and 6 μg protein in the reaction medium. The optimal activity was attained at 37 °C in a pH value of 8.0. The necessity of the cofactors Ca2+ and Mg2+ for the enzymatic activity was confirmed through a curve of concentration of 0-1000 µM in the reaction medium, with both cofactors showing similar effects in the enzymatic activity. The Chevillard plot revealed that the hydrolysis of eATP and eADP occurred at the same active site of the enzyme. The analyses of E-ATPDases inhibitor and enzyme specificity showed possible involvement of E-NTPDase isoforms - 1 and - 2 in the isolated cells. Furthermore, different kinetic behavior of the nucleotide hydrolysis in each resident subpopulation lymphocyte was observed in this study, as MLL showed the higher Vmax with the lowest km values, while TL had the lowest Vmax and high km values. The kinetic values for E-NTPDase activity of each immune tissue lymphocytes can be an important therapeutic target for numeral immune-related diseases.
Collapse
Affiliation(s)
- Pedro Henrique Doleski
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria, Brazil.
- Department of Microbiology and Parasitology, Universidade Federal de Santa Maria, Santa Maria, Brazil.
| | - Fernanda Licker Cabral
- Department of Microbiology and Parasitology, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Matheus Henrique Jantsch
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Renan Silva Ebone
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Stephen Adeniyi Adefegha
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria, Brazil
- Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Daniela Bitencourt Rosa Leal
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria, Brazil
- Department of Microbiology and Parasitology, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | | |
Collapse
|
10
|
Taylor CA, Glover M, Maher J. CAR-T cell technologies that interact with the tumour microenvironment in solid tumours. Expert Rev Clin Immunol 2024; 20:849-871. [PMID: 39021098 DOI: 10.1080/1744666x.2024.2380894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Chimeric antigen receptor (CAR) T-cells have emerged as a ground-breaking therapy for the treatment of hematological malignancies due to their capacity for rapid tumor-specific killing and long-lasting tumor immunity. However, the same success has not been observed in patients with solid tumors. Largely, this is due to the additional challenges imposed by safe and uniform target selection, inefficient CAR T-cell access to sites of disease and the presence of a hostile immunosuppressive tumor microenvironment. AREAS COVERED Literature was reviewed on the PubMed database from the first description of a CAR by Kuwana, Kurosawa and colleagues in December 1987 through to the present day. This literature indicates that in order to tackle solid tumors, CAR T-cells can be further engineered with additional armoring strategies that facilitate trafficking to and infiltration of malignant lesions together with reversal of suppressive immune checkpoints that operate within solid tumor lesions. EXPERT OPINION In this review, we describe a number of recent advances in CAR T-cell technology that set out to combat the problems imposed by solid tumors including tumor recruitment, infiltration, immunosuppression, metabolic compromise, and hypoxia.
Collapse
Affiliation(s)
| | | | - John Maher
- Leucid Bio Ltd, Guy's Hospital, London, UK
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, UK
- Department of Immunology, Eastbourne Hospital, Eastbourne, East Sussex, UK
| |
Collapse
|
11
|
Kaplinsky N, Williams K, Watkins D, Adams M, Stanbery L, Nemunaitis J. Regulatory role of CD39 and CD73 in tumor immunity. Future Oncol 2024; 20:1367-1380. [PMID: 38652041 PMCID: PMC11321403 DOI: 10.2217/fon-2023-0871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/07/2024] [Indexed: 04/25/2024] Open
Abstract
CD39 is the rate-limiting enzyme for the molecular signal cascade leading to the generation of ADP and adenosine monophosphate (AMP). In conjunction with CD73, CD39 converts adenosine triphosphate (ATP) to ADP and AMP, which leads to the accumulation of immunosuppressive adenosine in the tumor microenvironment. This review focuses on the role of CD39 and CD73 in immune response and malignant progression, including the expression of CD39 within the tumor microenvironment and its relationship to immune effector cells, and its role in antigen presentation. The role of CD39- and CD73-targeting therapeutics and cancer-directed clinical trials investigating CD39 modulation are also explored.
Collapse
Affiliation(s)
| | - Kada Williams
- University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Dean Watkins
- University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Molly Adams
- University of Toledo College of Medicine, Toledo, OH 43614, USA
| | | | | |
Collapse
|
12
|
Aoki T, Wong V, Yin T, Nakamura E, Endo Y, Hayashida K, Robson SC, Nandurkar H, Diamond B, Kim SJ, Murao A, Wang P, Becker LB, Shinozaki K. Immune cell expression patterns of CD39/CD73 ectonucleotidases in rodent models of cardiac arrest and resuscitation. Front Immunol 2024; 15:1362858. [PMID: 38545102 PMCID: PMC10967020 DOI: 10.3389/fimmu.2024.1362858] [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: 12/29/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Background Cardiac arrest (CA) is a significant public health concern. There is the high imminent mortality and survival in those who are resuscitated is substantively compromised by the post-CA syndrome (PCAS), characterized by multiorgan ischemia-reperfusion injury (IRI). The inflammatory response in PCAS is complex and involves various immune cell types, including lymphocytes and myeloid cells that have been shown to exacerbate organ IRI, such as myocardial infarction. Purinergic signaling, as regulated by CD39 and CD73, has emerged as centrally important in the context of organ-specific IRI. Hence, comprehensive understanding of such purinergic responses may be likewise imperative for improving outcomes in PCAS. Methods We have investigated alterations of immune cell populations after CA by utilizing rodent models of PCAS. Blood and spleen were collected after CA and resuscitation and underwent flow cytometry analysis to evaluate shifts in CD3+CD4+ helper T cells, CD3+CD8a+ cytotoxic T cells, and CD4/CD8a ratios. We then examined the expression of CD39 and CD73 across diverse cell types, including myeloid cells, T lymphocytes, and B lymphocytes. Results In both rat and mouse models, there were significant increases in the frequency of CD3+CD4+ T lymphocytes in PCAS (rat, P < 0.01; mouse, P < 0.001), with consequently elevated CD4/CD8a ratios in whole blood (both, P < 0.001). Moreover, CD39 and CD73 expression on blood leukocytes were markedly increased (rat, P < 0.05; mouse, P < 0.01 at 24h). Further analysis in the experimental mouse model revealed that CD11b+ myeloid cells, with significant increase in their population (P < 0.01), had high level of CD39 (88.80 ± 2.05 %) and increased expression of CD73 (P < 0.05). CD19+ B lymphocytes showed slight increases of CD39 (P < 0.05 at 2h) and CD73 (P < 0.05 at 2h), while, CD3+ T lymphocytes had decreased levels of them. These findings suggested a distinct patterns of expression of CD39 and CD73 in these specific immune cell populations after CA. Conclusions These data have provided comprehensive insights into the immune response after CA, highlighting high-level expressions of CD39 and CD73 in myeloid cells.
Collapse
Affiliation(s)
- Tomoaki Aoki
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Vanessa Wong
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- State University of New York Downstate Medical Center, NY, United States
| | - Tai Yin
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Eriko Nakamura
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Yusuke Endo
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Kei Hayashida
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Simon C. Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Harshal Nandurkar
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Betty Diamond
- Institutes of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Sun Jung Kim
- Institutes of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Lance B. Becker
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY, United States
| | - Koichiro Shinozaki
- Department of Emergency Med-Cardiopulmonary, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Institutes of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY, United States
- Department of Emergency & Critical Care Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
13
|
Zhang Y, Lu Y, Wang X, He K, Fang M, Xu J, Xu Y, Tao F, Lü P. Causal Relationship between Immune Cells and Gynecological Cancers through Bidirectional and Multivariable Mendelian Randomization Analyses. J Cancer 2024; 15:1880-1889. [PMID: 38434980 PMCID: PMC10905409 DOI: 10.7150/jca.92627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/27/2024] [Indexed: 03/05/2024] Open
Abstract
Background: Evidence suggests potential associations between gynecological malignancies and various immune cell chemicals and systems. However, establishing a causal relationship remains uncertain. Methods: This work employed Wald ratio for one single-nucleotide polymorphism (SNP) or the inverse-variance weighted method (IVW) for multiple SNPs to conduct bidirectional two-sample Mendelian randomization (MR) analysis by utilizing genome-wide association study (GWAS) data. We employed supplementary methods, including MR-Egger and weighted median methods, to detect and correct for the influence of horizontal pleiotropy. In addition, we also use colocalization analysis for further validation. Results: In IVW analysis, increases in relative count of circulating CD11c+ HLA-DR++ conventional dendritic cells (cDC) were associated with an elevated risk of breast cancer (OR [95% CI], 1.1295 [1.0632-1.2000], P = 8.044 × 10-5), while elevated levels of HLA-DR on plasmacytoid dendritic cells (DC) and HLA-DR on DC were protective against breast cancer. In addition, actual count of CD39+ resting Treg AC was also shown to be causally associated with the development of ovarian cancer, whereas a high relative count of CD28+ CD45RA- CD8+ T cells reduced the risk of cervical cancer. Sensitivity analysis revealed almost no evidence of bias in the current study. Multivariable MR (MVMR) analyses further confirmed a direct impact of the CD11c+ HLA-DR++ cDC immune phenotype on breast cancer. Colocalization analysis showed the lead SNP, rs780094, suggesting HLA-DR GWAS shared a common genetic mechanism with breast cancer. Conclusions: The MR study identified significant causal relationships between multiple immunophenotypes and breast cancer, aiming to provide clinicians with some reference for cancer prediction and explore further potential associations between immune phenotypes and gynecologic tumors.
Collapse
Affiliation(s)
- Yangyang Zhang
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China; Shanghai Medical College, Fudan University, Shanghai, China
| | - Yangyuxiao Lu
- The First Clinical Medical College, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| | - Xuanyu Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Keren He
- The First Clinical Medical College, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| | - Mengqi Fang
- The First Clinical Medical College, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| | - Jiabao Xu
- Department of Immunology and Microbiology, Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ye Xu
- Department of Immunology and Microbiology, Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Fangfang Tao
- Department of Immunology and Microbiology, Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ping Lü
- Department of TCM, Taizhou First People's Hospital, Taizhou, Zhejiang 318020, China
| |
Collapse
|
14
|
Zhang C, Wang K, Wang H. Adenosine in cancer immunotherapy: Taking off on a new plane. Biochim Biophys Acta Rev Cancer 2023; 1878:189005. [PMID: 37913941 DOI: 10.1016/j.bbcan.2023.189005] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
As a new pillar of cancer therapy, tumor immunotherapy has brought irreplaceable durable responses in tumors. Considering its low response rate, additional immune regulatory mechanisms will be critical for the development of next-generation immune therapeutics. As a key regulatory mechanism, adenosine (ADO) protects tissues from excessive immune responses, but as a metabolite highly concentrated in tumor microenvironments, extracellular adenosine acts on adenosine receptors (mainly A2A receptors) expressed on MDSCs, Tregs, NK cells, effector T cells, DCs, and macrophages to promote tumor cell escape from immune surveillance by inhibiting the immune response. Amounting preclinical studies have demonstrated the adenosine pathway as a novel checkpoint for immunotherapy. Large number of adenosine pathway targeting clinical trials are now underway, including antibodies against CD39 and CD73 as well as A2A receptor inhibitors. There has been evidence of antitumor efficacy of these inhibitors in early clinical trials among a variety of tumors such as breast cancer, prostate cancer, non-small cell lung cancer, etc. As more clinical trial results are published, the combination of blockade of this pathway with immune checkpoint inhibitors, targeted drugs, traditional chemotherapy medications, radiotherapy and endocrine therapy will provide cancer patients with better clinical outcomes. We would elaborate on the role of CD39-CD73-A2AR pathway in the contribution of tumor microenvironment and the targeting of the adenosinergic pathway for cancer therapy in the review.
Collapse
Affiliation(s)
- Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Kai Wang
- Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
15
|
Riaz F, Huang Z, Pan F. Targeting post-translational modifications of Foxp3: a new paradigm for regulatory T cell-specific therapy. Front Immunol 2023; 14:1280741. [PMID: 37936703 PMCID: PMC10626496 DOI: 10.3389/fimmu.2023.1280741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
A healthy immune system is pivotal for the hosts to resist external pathogens and maintain homeostasis; however, the immunosuppressive tumor microenvironment (TME) damages the anti-tumor immunity and promotes tumor progression, invasion, and metastasis. Recently, many studies have found that Foxp3+ regulatory T (Treg) cells are the major immunosuppressive cells that facilitate the formation of TME by promoting the development of various tumor-associated cells and suppressing the activity of effector immune cells. Considering the role of Tregs in tumor progression, it is pivotal to identify new therapeutic drugs to target and deplete Tregs in tumors. Although several studies have developed strategies for targeted deletion of Treg to reduce the TME and support the accumulation of effector T cells in tumors, Treg-targeted therapy systematically affects the Treg population and may lead to the progression of autoimmune diseases. It has been understood that, nevertheless, in disease conditions, Foxp3 undergoes several definite post-translational modifications (PTMs), including acetylation, glycosylation, phosphorylation, ubiquitylation, and methylation. These PTMs not only elevate or mitigate the transcriptional activity of Foxp3 but also affect the stability and immunosuppressive function of Tregs. Various studies have shown that pharmacological targeting of enzymes involved in PTMs can significantly influence the PTMs of Foxp3; thus, it may influence the progression of cancers and/or autoimmune diseases. Overall, this review will help researchers to understand the advances in the immune-suppressive mechanisms of Tregs, the post-translational regulations of Foxp3, and the potential therapeutic targets and strategies to target the Tregs in TME to improve anti-tumor immunity.
Collapse
Affiliation(s)
| | | | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
16
|
Klaver D, Thurnher M. P2Y 11/IL-1 receptor crosstalk controls macrophage inflammation: a novel target for anti-inflammatory strategies? Purinergic Signal 2023; 19:501-511. [PMID: 37016172 PMCID: PMC10073626 DOI: 10.1007/s11302-023-09932-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/06/2023] [Indexed: 04/06/2023] Open
Abstract
Although first cloning of the human ATP receptor P2Y11 was successful 25 years ago, the exact downstream signaling pathways of P2Y11 receptor, which can couple to Gq and Gs proteins, have remained unclear. Especially the lack of rodent models as well as the limited availability of antibodies and pharmacological tools have hampered examination of P2Y11 expression and function. Many meaningful observations related to P2Y11 have been made in primary immune cells, indicating that P2Y11 receptors are important regulators of inflammation and cell migration, also by controlling mitochondrial activity. Our recent studies have shown that P2Y11 is upregulated during macrophage development and activates signaling through IL-1 receptor, which is well known for its ability to direct inflammatory and migratory processes. This review summarizes the results of the first transcriptomic and secretomic analyses of both, ectopic and native P2Y11 receptors, and discusses how P2Y11 crosstalk with the IL-1 receptor may govern anti-inflammatory and pro-angiogenic processes in human M2 macrophages.
Collapse
Affiliation(s)
- Dominik Klaver
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innrain 66a, 6020 Innsbruck, Austria
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innrain 66a, 6020 Innsbruck, Austria
| |
Collapse
|
17
|
Seneviratne HK. Nucleoside Triphosphate Diphosphohydrolase 1 Exhibits Enzymatic Activity toward Tenofovir Diphosphate. Drug Metab Dispos 2023; 51:385-391. [PMID: 36396461 DOI: 10.1124/dmd.122.000855] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
Tenofovir (TFV; prescribed as TFV disoproxil fumarate and TFV alafenamide prodrugs) is currently used for HIV prevention and treatment. TFV must be phosphorylated twice into TFV-diphosphate (TFV-DP) to become pharmacologically active. Previously, we reported heterogeneity in TFV-DP distribution in colorectal tissue (a putative site of HIV infection) sections collected from research participants receiving a TFV-containing enema. This observed heterogeneity is likely multifactorial. Of note, TFV-DP is structurally similar to ATP. It is known that nucleotidases such as nucleoside triphosphate diphosphohydrolases (NTPDases) dephosphorylate ATP. Thus, it was hypothesized that NTPDase-mediated dephosphorylation plays a role in regulating TFV-DP levels in colorectal tissue. To test this hypothesis, recombinant NTPDase proteins (NTPDase 1, 3, 4, 5, 6, and 8) were incubated, individually, with TFV-DP to determine their abilities to dephosphorylate TFV-DP in vitro. Following incubations, TFV-DP dephosphorylation was determined using both malachite green phosphate assays and ultrahigh-performance liquid chromatography tandem mass spectrometry. From these, NTPDase 1 exhibited the highest activity toward TFV-DP. Further, enzyme kinetic analysis revealed Michaelis-Menten kinetics for NTPDase 1-mediated TFV-DP dephosphorylation. Next, immunoblot analyses were conducted to confirm the expression of NTPDase 1 protein in human colorectal tissue. Liquid chromatography coupled to mass spectrometry proteomics analysis was used to measure the relative abundance of NTPDases in human colorectal tissue among healthy adult individuals (n = 4). These analyses confirmed the high abundance of NTPDase 1 in human colorectal tissue. Taken together, results suggest that NTPDase 1 may contribute to the regulation of TFV-DP levels. The above data provide important insights into the dephosphorylation of TFV-DP. SIGNIFICANCE STATEMENT: Nucleoside triphosphate diphosphohydrolases (NTPDases) that are involved in enzymatic ATP dephosphorylation may contribute to tenofovir-diphosphate (TFV-DP) dephosphorylation, leading to its inactivation. In this study, the NTPDases responsible for TFV-DP dephosphorylation in vitro and their expression in human colorectal tissue were investigated. Through this work, it was demonstrated that NTPDase 1 has the highest activity toward TFV-DP dephosphorylation, and it was abundant in human colorectal tissue. Importantly, these studies will increase our understanding of TFV-DP disposition.
Collapse
Affiliation(s)
- Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County and Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
18
|
Bi C, Schäkel L, Mirza S, Sylvester K, Pelletier J, Lee SY, Pillaiyar T, Sévigny J, Müller CE. Synthesis and structure-activity relationships of ticlopidine derivatives and analogs as inhibitors of ectonucleotidase CD39. Bioorg Chem 2023; 135:106460. [PMID: 37023582 DOI: 10.1016/j.bioorg.2023.106460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023]
Abstract
Ticlopidine is an antithrombotic prodrug of the thienotetrahydropyridine family. For platelet inhibition it has to undergo oxidative ring-opening by cytochrome P450 enzymes. The resulting thiol reacts with a cysteine residue of the purinergic P2Y12 receptor on thrombocytes resulting in covalent receptor blockade. Ticlopidine in its intact, not-metabolized form was previously shown to inhibit ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1, also known as cluster of differentiation (CD) 39). CD39 catalyzes the extracellular hydrolysis of ATP via ADP to AMP, which is further hydrolyzed by ecto-5'-nucleotidase (CD73) to adenosine. CD39 inhibition has been proposed as a novel strategy to increase the extracellular concentration of antiproliferative ATP, while decreasing immunosuppressive and cancer-promoting adenosine levels. In the present study, we performed an extensive structure-activity relationship (SAR) analysis of ticlopidine derivatives and analogs as CD39 inhibitors followed by an in-depth characterization of selected compounds. Altogether 74 compounds were synthesized, 41 of which are new, not previously described in literature. Benzotetrahydropyridines, in which the metabolically labile thiophene is replaced by a benzene ring, were discovered as a new class of allosteric CD39 inhibitors.
Collapse
|
19
|
Brum da Silva Nunes V, Kehl Dias C, Nathali Scholl J, Nedel Sant'Ana A, de Fraga Dias A, Granero Farias M, Alegretti AP, Sosnoski M, Esteves Daudt L, Bohns Michalowski M, Oliveira Battastini AM, Paz AA, Figueiró F. Lymphocytes from B-acute lymphoblastic leukemia patients present differential regulation of the adenosinergic axis depending on risk stratification. Discov Oncol 2022; 13:143. [PMID: 36581667 PMCID: PMC9800668 DOI: 10.1007/s12672-022-00602-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Although risk-stratified chemotherapy regimens improve B-cell acute lymphoblastic leukemia (B-ALL) clinical outcome, relapse occurs in a significant number of cases. The identification of new therapeutic targets as well as prognostic and diagnostic biomarkers can improve B-ALL patients' clinical outcomes. Purinergic signaling is an important pathway in cancer progression, however the expression of ectonucleotidases and their impact on immune cells in B-ALL lacks exploration. We aimed to analyze the expression of ectonucleotidases in B-ALL patients' lymphocyte subpopulations. METHODS Peripheral blood samples from 15 patients diagnosed with B-ALL were analyzed. Flow cytometry was used to analyze cellularity, expression level of CD38, CD39, and CD73, and frequency of [Formula: see text], and [Formula: see text] in lymphocyte subpopulations. Plasma was used for cytokines (by CBA kit) and adenine nucleosides/nucleotides detection (by HPLC). RESULTS Comparing B-ALL patients to health donors, we observed an increase of CD4 + and CD8 + T-cells. In addition, a decrease in CD38 expression in B and Treg subpopulations and an increase in CD39+ CD73+ frequency in Breg and CD8+ T-cells. Analyzing cytokines and adenine nucleosides/nucleotides, we found a decrease in TNF, IL-1β, and ADO concentrations, together with an increase in AMP in B-ALL patients' plasma. CONCLUSION As immunomodulators, the expression of ectonucleotidases might be associated with activation states, as well as the abundance of different cellular subsets. We observed a pro-tumor immunity expression profile in B-ALL patients at diagnosis, being associated with cell exhaustion and immune evasion in B-ALL.
Collapse
Affiliation(s)
- Vitória Brum da Silva Nunes
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Camila Kehl Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Juliete Nathali Scholl
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Alexia Nedel Sant'Ana
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Amanda de Fraga Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | - Ana Paula Alegretti
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Monalisa Sosnoski
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Liane Esteves Daudt
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Mariana Bohns Michalowski
- Hospital de Clínicas de Porto Alegre/HCPA, Porto Alegre, RS, CEP 90035-903, Brazil
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Faculdade de Medicina, UFRGS, Porto Alegre, RS, 90035-003, Brazil
| | - Ana Maria Oliveira Battastini
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | - Fabrício Figueiró
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, CEP 90035-003, Brazil.
| |
Collapse
|
20
|
Willcox A, Lee NT, Nandurkar HH, Sashindranath M. CD39 in the development and progression of pulmonary arterial hypertension. Purinergic Signal 2022; 18:409-419. [PMID: 35947229 PMCID: PMC9832216 DOI: 10.1007/s11302-022-09889-9] [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: 04/25/2022] [Accepted: 07/21/2022] [Indexed: 01/14/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating progressive disease characterised by pulmonary arterial vasoconstriction and vascular remodelling. Endothelial dysfunction has emerged as a contributing factor in the development of PAH. However, despite progress in the understanding of the pathophysiology of this disease, current therapies fail to impact upon long-term outcomes which remain poor in most patients. Recent observations have suggested the disturbances in the balance between ATP and adenosine may be integral to the vascular remodelling seen in PAH. CD39 is an enzyme important in regulating these nucleos(t)ides which may also provide a novel pathway to target for future therapies. This review summarises the role of adenosine signalling in the development and progression of PAH and highlights the therapeutic potential of CD39 for treatment of PAH.
Collapse
Affiliation(s)
- Abbey Willcox
- Australian Centre for Blood Diseases, Central Clinical School, Monash University and Alfred Health, Monash AMREP Building, Level 1, Walkway, via The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
| | - Natasha Ting Lee
- Australian Centre for Blood Diseases, Central Clinical School, Monash University and Alfred Health, Monash AMREP Building, Level 1, Walkway, via The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Harshal H Nandurkar
- Australian Centre for Blood Diseases, Central Clinical School, Monash University and Alfred Health, Monash AMREP Building, Level 1, Walkway, via The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Maithili Sashindranath
- Australian Centre for Blood Diseases, Central Clinical School, Monash University and Alfred Health, Monash AMREP Building, Level 1, Walkway, via The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| |
Collapse
|
21
|
Dwyer KM. Burnstock oration - purinergic signalling in kidney transplantation. Purinergic Signal 2022; 18:387-393. [PMID: 35471483 PMCID: PMC9832191 DOI: 10.1007/s11302-022-09865-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/06/2022] [Indexed: 01/14/2023] Open
Abstract
Kidney transplantation is the preferred treatment for individuals with kidney failure offering improved quality and quantity of life. Despite significant advancements in short term graft survival, longer term survival rates have not improved greatly mediated in large by chronic antibody mediated rejection. Strategies to reduce the donor kidney antigenic load may translate to improved transplant survival. CD39 on the vascular endothelium and on circulating cells, in particular regulatory T cells (Treg), is upregulated in response to hypoxic stimuli and plays a critical role in regulating the immune response removing proinflammatory ATP and generating anti-inflammatory adenosine. Herein, the role of CD39 in reducing ischaemia-reperfusion injury (IRI) and on Treg within the context of kidney transplantation is reviewed.
Collapse
Affiliation(s)
- Karen M. Dwyer
- grid.1021.20000 0001 0526 7079School of Medicine, Deakin University, Geelong, 3220 Australia
| |
Collapse
|
22
|
Secreted immune metabolites that mediate immune cell communication and function. Trends Immunol 2022; 43:990-1005. [PMID: 36347788 DOI: 10.1016/j.it.2022.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
Metabolites are emerging as essential factors for the immune system that are involved in both metabolic circuits and signaling cascades. Accumulated evidence suggests that altered metabolic programs initiated by the activation and maturation of immune cell types are accompanied by the delivery of various metabolites into the local environment. We propose that, in addition to protein/peptide ligands, secreted immune metabolites (SIMets) are essential components of immune communication networks that fine-tune immune responses under homeostatic and pathological conditions. We summarize recent advances in our understanding of SIMets and discuss the potential mechanisms by which some metabolites engage in immunological responses through receptor-, transporter-, and post-translational-mediated regulation. These insights may contribute to understanding physiology and developing effective therapeutics for inflammatory and immune-mediated diseases.
Collapse
|
23
|
Impact of ROS-Dependent Lipid Metabolism on Psoriasis Pathophysiology. Int J Mol Sci 2022; 23:ijms232012137. [PMID: 36292991 PMCID: PMC9602909 DOI: 10.3390/ijms232012137] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
Psoriasis is the most common autoimmune disease, yet its pathophysiology is not fully understood. It is now believed that psoriasis is caused by the increased activation of immune cells, especially Th1 lymphocytes. However, in psoriasis, immune cells interfere with the metabolism of keratinocytes, leading to their increased activation. Therefore, the pathophysiology of psoriasis is currently associated with the overproduction of ROS, which are involved in the activation of immune cells and keratinocytes as well as the modulation of various signaling pathways within them. Nevertheless, ROS modulate the immune system by also boosting the increasing generation of various lipid mediators, such as products of lipid peroxidation as well as endocannabinoids and prostaglandins. In psoriasis, the excessive generation of ROS and lipid mediators is observed in different immune cells, such as granulocytes, dendritic cells, and lymphocytes. All of the above may be activated by ROS and lipid mediators, which leads to inflammation. Nevertheless, ROS and lipid mediators regulate lymphocyte differentiation in favor of Th1 and may also interact directly with keratinocytes, which is also observed in psoriasis. Thus, the analysis of the influence of oxidative stress and its consequences for metabolic changes, including lipidomic ones, in psoriasis may be of diagnostic and therapeutic importance.
Collapse
|
24
|
Hall BM, Verma ND, Tran GT, Hodgkinson SJ. Transplant Tolerance, Not Only Clonal Deletion. Front Immunol 2022; 13:810798. [PMID: 35529847 PMCID: PMC9069565 DOI: 10.3389/fimmu.2022.810798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The quest to understand how allogeneic transplanted tissue is not rejected and how tolerance is induced led to fundamental concepts in immunology. First, we review the research that led to the Clonal Deletion theory in the late 1950s that has since dominated the field of immunology and transplantation. At that time many basic mechanisms of immune response were unknown, including the role of lymphocytes and T cells in rejection. These original observations are reassessed by considering T regulatory cells that are produced by thymus of neonates to prevent autoimmunity. Second, we review "operational tolerance" induced in adult rodents and larger animals such as pigs. This can occur spontaneously especially with liver allografts, but also can develop after short courses of a variety of rejection inhibiting therapies. Over time these animals develop alloantigen specific tolerance to the graft but retain the capacity to reject third-party grafts. These animals have a "split tolerance" as peripheral lymphocytes from these animals respond to donor alloantigen in graft versus host assays and in mixed lymphocyte cultures, indicating there is no clonal deletion. Investigation of this phenomenon excludes many mechanisms, including anti-donor antibody blocking rejection as well as anti-idiotypic responses mediated by antibody or T cells. This split tolerance is transferred to a second immune-depleted host by T cells that retain the capacity to effect rejection of third-party grafts by the same host. Third, we review research on alloantigen specific inhibitory T cells that led to the first identification of the CD4+CD25+T regulatory cell. The key role of T cell derived cytokines, other than IL-2, in promoting survival and expansion of antigen specific T regulatory cells that mediate transplant tolerance is reviewed. The precise methods for inducing and diagnosing operational tolerance remain to be defined, but antigen specific T regulatory cells are key mediators.
Collapse
Affiliation(s)
- Bruce M. Hall
- Immune Tolerance Laboratory, School of Medicine, University of New South Wales (UNSW) Sydney, Ingham Institute, and Renal Service and Multiple Sclerosis Clinic, Liverpool Hospital, Liverpool, NSW, Australia
| | | | | | | |
Collapse
|
25
|
Sun C, Wang B, Hao S. Adenosine-A2A Receptor Pathway in Cancer Immunotherapy. Front Immunol 2022; 13:837230. [PMID: 35386701 PMCID: PMC8977492 DOI: 10.3389/fimmu.2022.837230] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/28/2022] [Indexed: 12/14/2022] Open
Abstract
A2A receptors (A2AR), a typical GPCR with a high affinity for adenosine, was expressed in many immune cells, such as regulatory T cells, cytotoxic T cells, macrophages, etc. Adenosine binding to the A2AR receptor activates the typical G protein and triggers the cAMP/PKA/CREB pathway. The adenosine-A2AR pathway plays an important role in protecting normal organs and tissues from the autoimmune response of immune cells. However, many solid tumors hijack the adenosine-A2AR pathway by promoting adenosine accumulation. The activation of the A2AR pathway inhibited the immune response of immune cells and then promotes the immune escape of tumor cells in the tumor microenvironment. Recently, both animal experiments and clinical trials indicated that blocking the adenosine pathway can inhibit the progression of a variety of solid tumors. In addition, it is encouraging that A2AR blockade combined with CAR T cells therapy showed better anti-tumor efficacy. Therefore, this review will discuss the role of the adenosine-A2AR pathway in the tumor microenvironment and summarize recent advances of A2AR-cancer related studies.
Collapse
Affiliation(s)
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
26
|
Marginal Zone B-Cell Populations and Their Regulatory Potential in the Context of HIV and Other Chronic Inflammatory Conditions. Int J Mol Sci 2022; 23:ijms23063372. [PMID: 35328792 PMCID: PMC8949885 DOI: 10.3390/ijms23063372] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Inflammation in the context of Human Immunodeficiency Virus (HIV) establishes early and persists beyond antiretroviral therapy (ART). As such, we have shown excess B-cell activating factor (BAFF) in the blood of HIV-infected progressors, as soon as in the acute phase, and despite successful ART. Excess BAFF was associated with deregulation of the B-cell compartment; notably, with increased frequencies of a population sharing features of both transitional immature (TI) and marginal zone (MZ) B-cells, we termed Marginal Zone precursor-like (MZp). We have reported similar observations with HIV-transgenic mice, Simian Immunodeficiency Virus (SIV)-infected macaques, and more recently, with HIV-infected Beninese commercial sex workers, which suggests that excess BAFF and increased frequencies of MZp B-cells are reliable markers of inflammation in the context of HIV. Importantly, we have recently shown that in healthy individuals, MZps present an important regulatory B-cell (Breg) profile and function. Herein, we wish to review our current knowledge on MZ B-cell populations, especially their Breg status, and that of other B-cell populations sharing similar features. BAFF and its analog A Proliferation-Inducing Ligand (APRIL) are important in shaping the MZ B-cell pool; moreover, the impact that excess BAFF—encountered in the context of HIV and several chronic inflammatory conditions—may exert on MZ B-cell populations, Breg and antibody producing capacities is a threat to the self-integrity of their antibody responses and immune surveillance functions. As such, deregulations of MZ B-cell populations contribute to autoimmune manifestations and the development of MZ lymphomas (MZLs) in the context of HIV and other inflammatory diseases. Therefore, further comprehending the mechanisms regulating MZ B-cell populations and their functions could be beneficial to innovative therapeutic avenues that could be deployed to restore MZ B-cell immune competence in the context of chronic inflammation involving excess BAFF.
Collapse
|
27
|
The Ecto-5
′
nucleotidase/CD73 Mediates Leishmania amazonensis Survival in Macrophages. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9928362. [PMID: 35187176 PMCID: PMC8856795 DOI: 10.1155/2022/9928362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/30/2021] [Accepted: 12/28/2021] [Indexed: 11/23/2022]
Abstract
Endogenous nucleotides produced by various group of cells under inflammatory conditions act as potential danger signals in vivo. Extracellularly released nucleotides such as ATP are rapidly hydrolyzed to adenosine by the coordinated ectonucleotidase activities of CD39 and CD73. Leishmania is an obligate intracellular parasite of macrophages and capable of modulating host immune response in order to survive and multiply within host cells. In this study, the activity of CD73 induced by Leishmania amazonensis in infected macrophages has been investigated and correlated with parasite survival and infection in vitro. For this, the expression of CD39 and CD73, by flow cytometry, in murine peritoneal macrophages infected with metacyclic promastigotes of L. amazonensis has been analyzed. Our results showed that L. amazonensis-infected macrophages, unlike LPS-treated macrophages, increased CD73 expression. It was also noted that when CD73 enzymatic activity was blocked by α, β-methyleneadenosine 5′-diphosphate sodium salt (APCP), macrophage parasitism was significantly decreased. Interestingly, these effects were not associated with the production of TNF-α, IL-10, or nitric oxide (NO). Together, these data demonstrate that L. amazonensis induces a regulatory phenotype in macrophages, which by activating the CD39/CD73 pathway allows parasite survival through the action of immunomodulatory adenosine receptors.
Collapse
|
28
|
Eberhardt N, Bergero G, Mazzocco Mariotta YL, Aoki MP. Purinergic modulation of the immune response to infections. Purinergic Signal 2022; 18:93-113. [PMID: 34997903 PMCID: PMC8742569 DOI: 10.1007/s11302-021-09838-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023] Open
Abstract
Infectious diseases are caused by the invasion of pathogenic microorganisms such as fungi, bacteria, viruses, and parasites. After infection, disease progression relies on the complex interplay between the host immune response and the microorganism evasion strategies. The host's survival depends on its ability to mount an efficient protective anti-microbial response to accomplish pathogen clearance while simultaneously preventing tissue injury by keeping under control the excessive inflammatory process. The purinergic system has the dual function of regulating the immune response and triggering effector antimicrobial mechanisms. This review provides an overview of the current knowledge of the modulation of innate and adaptive immunity driven by the purinergic system during parasitic, bacterial and viral infections.
Collapse
Affiliation(s)
- Natalia Eberhardt
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET) - Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Present Address: Department of Medicine, Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, USA
| | - Gastón Bergero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET) - Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Yanina L. Mazzocco Mariotta
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET) - Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M. Pilar Aoki
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET) - Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Haya de La Torre and Medina Allende, Ciudad Universitaria, CP 5000 Córdoba, Argentina
| |
Collapse
|
29
|
Zhong SR, Kuang Q, Zhang F, Chen B, Zhong ZG. Functional roles of the microbiota-gut-brain axis in Alzheimer’s disease: Implications of gut microbiota-targeted therapy. Transl Neurosci 2021; 12:581-600. [PMID: 35070442 PMCID: PMC8724360 DOI: 10.1515/tnsci-2020-0206] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing scientific evidence demonstrates that the gut microbiota influences normal physiological homeostasis and contributes to pathogenesis, ranging from obesity to neurodegenerative diseases, such as Alzheimer’s disease (AD). Gut microbiota can interact with the central nervous system (CNS) through the microbiota-gut-brain axis. The interaction is mediated by microbial secretions, metabolic interventions, and neural stimulation. Here, we review and summarize the regulatory pathways (immune, neural, neuroendocrine, or metabolic systems) in the microbiota-gut-brain axis in AD pathogenesis. Besides, we highlight the significant roles of the intestinal epithelial barrier and blood–brain barrier (BBB) in the microbiota-gut-brain axis. During the progression of AD, there is a gradual shift in the gut microbiota and host co-metabolic relationship, leading to gut dysbiosis, and the imbalance of microbial secretions and metabolites, such as lipopolysaccharides (LPS) and short-chain fatty acids (SCFAs). These products may affect the CNS metabolic state and immune balance through the microbiota-gut-brain axis. Further, we summarize the potential microbiota-gut-brain axis-targeted therapy including carbohydrates, probiotics, dietary measures, and propose new strategies toward the development of anti-AD drugs. Taken together, the data in this review suggest that remodeling the gut microbiota may present a tractable strategy in the management and development of new therapeutics against AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Si-Ran Zhong
- School of Health Medicine, Guangzhou Huashang College , Guangzhou , 511300 , People’s Republic of China
| | - Qi Kuang
- School of Health Medicine, Guangzhou Huashang College , Guangzhou , 511300 , People’s Republic of China
| | - Fan Zhang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine , Guangzhou , 510006 , People’s Republic of China
| | - Ben Chen
- Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine , Nanning City , 530200, Guangxi Zhuang Autonomous Region , People’s Republic of China
| | - Zhen-Guo Zhong
- Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine , Nanning City , 530200, Guangxi Zhuang Autonomous Region , People’s Republic of China
| |
Collapse
|
30
|
Hall BM, Hall RM, Tran GT, Robinson CM, Wilcox PL, Rakesh PK, Wang C, Sharland AF, Verma ND, Hodgkinson SJ. Interleukin-5 (IL-5) Therapy Prevents Allograft Rejection by Promoting CD4 +CD25 + Ts2 Regulatory Cells That Are Antigen-Specific and Express IL-5 Receptor. Front Immunol 2021; 12:714838. [PMID: 34912327 PMCID: PMC8667344 DOI: 10.3389/fimmu.2021.714838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/01/2021] [Indexed: 12/26/2022] Open
Abstract
CD4+CD25+Foxp3+T cell population is heterogenous and contains three major sub-groups. First, thymus derived T regulatory cells (tTreg) that are naïve/resting. Second, activated/memory Treg that are produced by activation of tTreg by antigen and cytokines. Third, effector lineage CD4+CD25+T cells generated from CD4+CD25- T cells' activation by antigen to transiently express CD25 and Foxp3. We have shown that freshly isolated CD4+CD25+T cells are activated by specific alloantigen and IL-4, not IL-2, to Ts2 cells that express the IL-5 receptor alpha. Ts2 cells are more potent than naïve/resting tTreg in suppressing specific alloimmunity. Here, we showed rIL-5 promoted further activation of Ts2 cells to Th2-like Treg, that expressed foxp3, irf4, gata3 and il5. In vivo, we studied the effects of rIL-5 treatment on Lewis heart allograft survival in F344 rats. Host CD4+CD25+T cells were assessed by FACS, in mixed lymphocyte culture and by RT-PCR to examine mRNA of Ts2 or Th2-like Treg markers. rIL-5 treatment given 7 days after transplantation reduced the severity of rejection and all grafts survived ≥60d whereas sham treated rats fully rejected by day 31 (p<0.01). Treatment with anti-CD25 or anti-IL-4 monoclonal antibody abolished the benefits of treatment with rIL-5 and accelerated rejection. After 10d treatment with rIL-5, hosts' CD4+CD25+ cells expressed more Il5ra and responded to specific donor Lewis but not self. Enriched CD4+CD25+ cells from rIL-5 treated rats with allografts surviving >60 days proliferated to specific donor only when rIL-5 was present and did not proliferate to self or third party. These cells had more mRNA for molecules expressed by Th2-like Treg including Irf4, gata3 and Il5. These findings were consistent with IL-5 treatment preventing rejection by activation of Ts2 cells and Th2-like Treg.
Collapse
Affiliation(s)
- Bruce M Hall
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Rachael M Hall
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Giang T Tran
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Catherine M Robinson
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Paul L Wilcox
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Prateek K Rakesh
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Chuanmin Wang
- Transplantation Immunobiology Group, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra F Sharland
- Transplantation Immunobiology Group, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Nirupama D Verma
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| | - Suzanne J Hodgkinson
- Immune Tolerance Laboratory, South West Clinical School, University of New South Wales (UNSW) Sydney, Liverpool, NSW, Australia.,Ingham Institute of Applied Medical Research, Liverpool Hospital, Liverpool, NSW, Australia
| |
Collapse
|
31
|
Nascimento DC, Viacava PR, Ferreira RG, Damaceno MA, Piñeros AR, Melo PH, Donate PB, Toller-Kawahisa JE, Zoppi D, Veras FP, Peres RS, Menezes-Silva L, Caetité D, Oliveira AER, Castro ÍMS, Kauffenstein G, Nakaya HI, Borges MC, Zamboni DS, Fonseca DM, Paschoal JAR, Cunha TM, Quesniaux V, Linden J, Cunha FQ, Ryffel B, Alves-Filho JC. Sepsis expands a CD39 + plasmablast population that promotes immunosuppression via adenosine-mediated inhibition of macrophage antimicrobial activity. Immunity 2021; 54:2024-2041.e8. [PMID: 34473957 DOI: 10.1016/j.immuni.2021.08.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/14/2020] [Accepted: 08/06/2021] [Indexed: 12/17/2022]
Abstract
Sepsis results in elevated adenosine in circulation. Extracellular adenosine triggers immunosuppressive signaling via the A2a receptor (A2aR). Sepsis survivors develop persistent immunosuppression with increased risk of recurrent infections. We utilized the cecal ligation and puncture (CLP) model of sepsis and subsequent infection to assess the role of adenosine in post-sepsis immune suppression. A2aR-deficient mice showed improved resistance to post-sepsis infections. Sepsis expanded a subset of CD39hi B cells and elevated extracellular adenosine, which was absent in mice lacking CD39-expressing B cells. Sepsis-surviving B cell-deficient mice were more resistant to secondary infections. Mechanistically, metabolic reprogramming of septic B cells increased production of ATP, which was converted into adenosine by CD39 on plasmablasts. Adenosine signaling via A2aR impaired macrophage bactericidal activity and enhanced interleukin-10 production. Septic individuals exhibited expanded CD39hi plasmablasts and adenosine accumulation. Our study reveals CD39hi plasmablasts and adenosine as important drivers of sepsis-induced immunosuppression with relevance in human disease.
Collapse
Affiliation(s)
- Daniele Carvalho Nascimento
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; CNRS, UMR7355, Orleans, 45071, France.
| | - Paula Ramos Viacava
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Raphael Gomes Ferreira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Marina Alves Damaceno
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Annie Rocío Piñeros
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Paulo Henrique Melo
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Paula Barbim Donate
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Juliana Escher Toller-Kawahisa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Daniel Zoppi
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Flávio Protásio Veras
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Raphael Sanches Peres
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Luísa Menezes-Silva
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Diego Caetité
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Antonio Edson Rocha Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Ícaro Maia Santos Castro
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Gilles Kauffenstein
- UMR INSERM 1260, Regenerative NanoMedicine, University of Strasbourg, Strasbourg 60026, France
| | | | - Marcos Carvalho Borges
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Dario Simões Zamboni
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Denise Morais Fonseca
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Jonas Augusto Rizzato Paschoal
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Valerie Quesniaux
- CNRS, UMR7355, Orleans, 45071, France; Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans 45071, France
| | - Joel Linden
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Fernando Queíroz Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Bernhard Ryffel
- CNRS, UMR7355, Orleans, 45071, France; Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans 45071, France
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil.
| |
Collapse
|
32
|
Murtaza A, Afzal S, Zaman G, Saeed A, Pelletier J, Sévigny J, Iqbal J, Hassan A. Divergent synthesis and elaboration of structure activity relationship for quinoline derivatives as highly selective NTPDase inhibitor. Bioorg Chem 2021; 115:105240. [PMID: 34416508 DOI: 10.1016/j.bioorg.2021.105240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/25/2021] [Accepted: 08/02/2021] [Indexed: 01/11/2023]
Abstract
Quinoline derivatives have interesting biological profile. In continuation for the comprehensive evaluations of substituted quinoline derivatives against human nucleoside triphosphate diphosphohydrolases (h-NTPDases) a series of substituted quinoline derivatives (2a-g, 3a-f, 4, 5a-c, 6) was synthesized. The inhibitory activities of the synthesized compounds were evaluated against four isoenzymes of human nucleoside triphosphate diphosphohydrolases (h-NTPDases). These quinoline derivatives had IC50 (µM) values in the range of 0.20-1.75, 0.77-2.20, 0.36-5.50 and 0.90-1.82 for NTPDase1, NTPDase2, NTPDase3 and NTPDase8, respectively. The derivative 3f was the most active compound against NTPDase1 (IC50, 0.20 ± 0.02 µM) that also possessed selectivity towards NTPDase1. Similarly, derivative 3b (IC50, 0.77 ± 0.06), 2h (IC50, 0.36 ± 0.01) and 2c (IC50, 0.90 ± 0.08) displayed excellent activity corresponding to NTPDase2, NTPDase3 and NTPdase8. The compound 5c emerged as a selective inhibitor of NTPDase8. The most active compounds were then investigated to determine their mode of inhibition and finally binding interactions of the active compounds were analyzed through molecular docking studies. The obtained results strongly support the quinoline scaffold's potential as potent and selective NTPDase inhibitor.
Collapse
Affiliation(s)
- Amna Murtaza
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Saira Afzal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Gohar Zaman
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Julie Pelletier
- Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Jean Sévigny
- Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada; Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, QC, Canada
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan.
| | - Abbas Hassan
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
33
|
Medina CB, Chiu YH, Stremska ME, Lucas CD, Poon I, Tung KS, Elliott MR, Desai B, Lorenz UM, Bayliss DA, Ravichandran KS. Pannexin 1 channels facilitate communication between T cells to restrict the severity of airway inflammation. Immunity 2021; 54:1715-1727.e7. [PMID: 34283971 PMCID: PMC8363584 DOI: 10.1016/j.immuni.2021.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 01/14/2023]
Abstract
Allergic airway inflammation is driven by type-2 CD4+ T cell inflammatory responses. We uncover an immunoregulatory role for the nucleotide release channel, Panx1, in T cell crosstalk during airway disease. Inverse correlations between Panx1 and asthmatics and our mouse models revealed the necessity, specificity, and sufficiency of Panx1 in T cells to restrict inflammation. Global Panx1-/- mice experienced exacerbated airway inflammation, and T-cell-specific deletion phenocopied Panx1-/- mice. A transgenic designed to re-express Panx1 in T cells reversed disease severity in global Panx1-/- mice. Panx1 activation occurred in pro-inflammatory T effector (Teff) and inhibitory T regulatory (Treg) cells and mediated the extracellular-nucleotide-based Treg-Teff crosstalk required for suppression of Teff cell proliferation. Mechanistic studies identified a Salt-inducible kinase-dependent phosphorylation of Panx1 serine 205 important for channel activation. A genetically targeted mouse expressing non-phosphorylatable Panx1S205A phenocopied the exacerbated inflammation in Panx1-/- mice. These data identify Panx1-dependent Treg:Teff cell communication in restricting airway disease.
Collapse
Affiliation(s)
- Christopher B Medina
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yu-Hsin Chiu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu, 30071, Taiwan
| | - Marta E Stremska
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Christopher D Lucas
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA
| | - Ivan Poon
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, VIC 3083, Australia
| | - Kenneth S Tung
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Michael R Elliott
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Bimal Desai
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ulrike M Lorenz
- Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908, USA; Departments of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA; VIB/UGent Inflammation Research Centre and the Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
34
|
Baghbani E, Noorolyai S, Shanehbandi D, Mokhtarzadeh A, Aghebati-Maleki L, Shahgoli VK, Brunetti O, Rahmani S, Shadbad MA, Baghbanzadeh A, Silvestris N, Baradaran B. Regulation of immune responses through CD39 and CD73 in cancer: Novel checkpoints. Life Sci 2021; 282:119826. [PMID: 34265363 DOI: 10.1016/j.lfs.2021.119826] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/22/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
The immunosuppressive tumor microenvironment has been implicated in attenuating anti-tumoral immune responses and tumor growth in various cancers. Inhibitory immune checkpoints have been introduced as the primary culprits for developing the immunosuppressive tumor microenvironment. Therefore, a better understanding of the cross-talk between inhibitory immune checkpoints in the tumor microenvironment can pave the way for introducing novel approaches for treating affected patients. Growing evidence indicates that CD39 and CD73, as novel checkpoints, can transform adenosine triphosphate (ATP)-mediated pro-inflammatory tumor microenvironment into an adenosine-mediated immunosuppressive one via the purinergic signaling pathway. Indeed, enzymatic processes of CD39 and CD73 have crucial roles in adjusting the extent, intensity, and chemical properties of purinergic signals. This study aims to review the biological function of CD39 and CD73 and shed light on their significance in regulating anti-tumoral immune responses in various cancers.
Collapse
Affiliation(s)
- Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vahid Khaze Shahgoli
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Cancer and Inflammation Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Shima Rahmani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nicola Silvestris
- IRCCS Bari, Italy, Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO, University of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
35
|
Role of Purinergic Signalling in Endothelial Dysfunction and Thrombo-Inflammation in Ischaemic Stroke and Cerebral Small Vessel Disease. Biomolecules 2021; 11:biom11070994. [PMID: 34356618 PMCID: PMC8301873 DOI: 10.3390/biom11070994] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/20/2022] Open
Abstract
The cerebral endothelium is an active interface between blood and the central nervous system. In addition to being a physical barrier between the blood and the brain, the endothelium also actively regulates metabolic homeostasis, vascular tone and permeability, coagulation, and movement of immune cells. Being part of the blood–brain barrier, endothelial cells of the brain have specialized morphology, physiology, and phenotypes due to their unique microenvironment. Known cardiovascular risk factors facilitate cerebral endothelial dysfunction, leading to impaired vasodilation, an aggravated inflammatory response, as well as increased oxidative stress and vascular proliferation. This culminates in the thrombo-inflammatory response, an underlying cause of ischemic stroke and cerebral small vessel disease (CSVD). These events are further exacerbated when blood flow is returned to the brain after a period of ischemia, a phenomenon termed ischemia-reperfusion injury. Purinergic signaling is an endogenous molecular pathway in which the enzymes CD39 and CD73 catabolize extracellular adenosine triphosphate (eATP) to adenosine. After ischemia and CSVD, eATP is released from dying neurons as a damage molecule, triggering thrombosis and inflammation. In contrast, adenosine is anti-thrombotic, protects against oxidative stress, and suppresses the immune response. Evidently, therapies that promote adenosine generation or boost CD39 activity at the site of endothelial injury have promising benefits in the context of atherothrombotic stroke and can be extended to current CSVD known pathomechanisms. Here, we have reviewed the rationale and benefits of CD39 and CD39 therapies to treat endothelial dysfunction in the brain.
Collapse
|
36
|
Haas CB, Lovászi M, Braganhol E, Pacher P, Haskó G. Ectonucleotidases in Inflammation, Immunity, and Cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:1983-1990. [PMID: 33879578 PMCID: PMC10037530 DOI: 10.4049/jimmunol.2001342] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/02/2021] [Indexed: 12/15/2022]
Abstract
Nucleoside triphosphate diphosphohydrolases (NTPDases) are a family of enzymes that hydrolyze nucleotides such as ATP, UTP, ADP, and UDP to monophosphates derivates such as AMP and UMP. The NTPDase family consists of eight enzymes, of which NTPDases 1, 2, 3, and 8 are expressed on cell membranes thereby hydrolyzing extracellular nucleotides. Cell membrane NTPDases are expressed in all tissues, in which they regulate essential physiological tissue functions such as development, blood flow, hormone secretion, and neurotransmitter release. They do so by modulating nucleotide-mediated purinergic signaling through P2 purinergic receptors. NTPDases 1, 2, 3, and 8 also play a key role during infection, inflammation, injury, and cancer. Under these conditions, NTPDases can contribute and control the pathophysiology of infectious, inflammatory diseases and cancer. In this review, we discuss the role of NTPDases, focusing on the less understood NTPDases 2-8, in regulating inflammation and immunity during infectious, inflammatory diseases, and cancer.
Collapse
Affiliation(s)
| | | | - Elizandra Braganhol
- Departamento de Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; and
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/National Institute of Alcohol Abuse and Alcoholism, Bethesda, MD
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY;
| |
Collapse
|
37
|
Maier-Begandt D, Comstra HS, Molina SA, Krüger N, Ruddiman CA, Chen YL, Chen X, Biwer LA, Johnstone SR, Lohman AW, Good ME, DeLalio LJ, Hong K, Bacon HM, Yan Z, Sonkusare SK, Koval M, Isakson BE. A venous-specific purinergic signaling cascade initiated by Pannexin 1 regulates TNFα-induced increases in endothelial permeability. Sci Signal 2021; 14:14/672/eaba2940. [PMID: 33653920 PMCID: PMC8011850 DOI: 10.1126/scisignal.aba2940] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The endothelial cell barrier regulates the passage of fluid between the bloodstream and underlying tissues, and barrier function impairment exacerbates the severity of inflammatory insults. To understand how inflammation alters vessel permeability, we studied the effects of the proinflammatory cytokine TNFα on transendothelial permeability and electrophysiology in ex vivo murine veins and arteries. We found that TNFα specifically decreased the barrier function of venous endothelium without affecting that of arterial endothelium. On the basis of RNA expression profiling and protein analysis, we found that claudin-11 (CLDN11) was the predominant claudin in venous endothelial cells and that there was little, if any, CLDN11 in arterial endothelial cells. Consistent with a difference in claudin composition, TNFα increased the permselectivity of Cl- over Na+ in venous but not arterial endothelium. The vein-specific effects of TNFα also required the activation of Pannexin 1 (Panx1) channels and the CD39-mediated hydrolysis of ATP to adenosine, which subsequently stimulated A2A adenosine receptors. Moreover, the increase in vein permeability required the activation of the Ca2+ channel TRPV4 downstream of Panx1 activation. Panx1-deficient mice resisted the pathologic effects of sepsis induced by cecal ligation and puncture on life span and lung vascular permeability. These data provide a targetable pathway with the potential to promote vein barrier function and prevent the deleterious effects of vascular leak in response to inflammation.
Collapse
Affiliation(s)
- Daniela Maier-Begandt
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.,Walter Brendel Center of Experimental Medicine, University Hospital, and Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Heather Skye Comstra
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Samuel A Molina
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nenja Krüger
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.,Institute of Animal Developmental and Molecular Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Xiaobin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Lauren A Biwer
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research, Virginia Tech, Roanoke, VA 24016, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060, USA
| | - Alexander W Lohman
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Leon J DeLalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Kwangseok Hong
- Department of Physical Education, College of Education, Chung-Ang University, Seoul 06974, South Korea
| | - Hannah M Bacon
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Zhen Yan
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA. .,Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA. .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
38
|
Zimmermann H. Ectonucleoside triphosphate diphosphohydrolases and ecto-5'-nucleotidase in purinergic signaling: how the field developed and where we are now. Purinergic Signal 2021; 17:117-125. [PMID: 33336318 PMCID: PMC7954995 DOI: 10.1007/s11302-020-09755-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022] Open
Abstract
Geoffrey Burnstock will be remembered as the scientist who set up an entirely new field of intercellular communication, signaling via nucleotides. The signaling cascades involved in purinergic signaling include intracellular storage of nucleotides, nucleotide release, extracellular hydrolysis, and the effect of the released compounds or their hydrolysis products on target tissues via specific receptor systems. In this context ectonucleotidases play several roles. They inactivate released and physiologically active nucleotides, produce physiologically active hydrolysis products, and facilitate nucleoside recycling. This review briefly highlights the development of our knowledge of two types of enzymes involved in extracellular nucleotide hydrolysis and thus purinergic signaling, the ectonucleoside triphosphate diphosphohydrolases, and ecto-5'-nucleotidase.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Goethe University, Institute of Cell Biology and Neuroscience, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
39
|
Naranjo-Gomez M, Cahen M, Lambour J, Boyer-Clavel M, Pelegrin M. Immunomodulatory Role of NK Cells during Antiviral Antibody Therapy. Vaccines (Basel) 2021; 9:137. [PMID: 33567792 PMCID: PMC7914599 DOI: 10.3390/vaccines9020137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Monoclonal antibodies (mAbs) are now considered as a therapeutic approach to prevent and treat severe viral infections. Using a mouse retroviral model, we showed that mAbs induce protective immunity (vaccinal effects). Here, we investigated the role of natural killer (NK) cells on this effect. NK cells are effector cells that are crucial to control viral propagation upon mAb treatment. However, their immunomodulatory activity during antiviral mAb immunotherapies has been little studied. Our data reveal that the mAb treatment of infected mice preserves the functional activation of NK cells. Importantly, functional NK cells play an essential role in preventing immune dysfunction and inducing antiviral protective immunity upon mAb therapy. Thus, NK cell depletion in mAb-treated, viral-infected mice leads to the upregulation of molecules involved in immunosuppressive pathways (i.e., PD-1, PD-L1 and CD39) on dendritic cells and T cells. NK cell depletion also abrogates the vaccinal effects induced by mAb therapy. Our data also reveal a role for IFNγ-producing NK cells in the enhancement of the B-cell responses through the potentiation of the B-cell helper properties of neutrophils. These findings suggest that preserved NK cell functions and counts might be required for achieving mAb-induced protective immunity. They open new prospects for improving antiviral immunotherapies.
Collapse
Affiliation(s)
- Mar Naranjo-Gomez
- IGMM, Univ Montpellier, CNRS, Montpellier, France; (M.N.-G.); (M.C.); (J.L.)
| | - Marine Cahen
- IGMM, Univ Montpellier, CNRS, Montpellier, France; (M.N.-G.); (M.C.); (J.L.)
| | - Jennifer Lambour
- IGMM, Univ Montpellier, CNRS, Montpellier, France; (M.N.-G.); (M.C.); (J.L.)
| | - Myriam Boyer-Clavel
- Montpellier Ressources Imagerie, Biocampus, Univ Montpellier, CNRS, Montpellier, France;
| | - Mireia Pelegrin
- IGMM, Univ Montpellier, CNRS, Montpellier, France; (M.N.-G.); (M.C.); (J.L.)
| |
Collapse
|
40
|
Yuan X, Li Y, Zhang AZ, Jiang CH, Li FP, Xie YF, Li JF, Liang WH, Zhang HJ, Liu CX, Pang LJ, Shen XH, Li F, Hu JM. Tumor-associated macrophage polarization promotes the progression of esophageal carcinoma. Aging (Albany NY) 2020; 13:2049-2072. [PMID: 33323552 PMCID: PMC7880404 DOI: 10.18632/aging.202201] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/22/2020] [Indexed: 12/14/2022]
Abstract
The immune response facilitated by tumor-associated macrophages is a vital determinant of tumor progression. We identified differentially expressed genes between various macrophage phenotypes in the Gene Expression Omnibus, and used Kaplan-Meier Plotter to determine which of them altered the prognosis of esophageal carcinoma patients. Fibrinogen-like protein 2 (FGL2), an immunosuppressive factor in the tumor microenvironment of various cancers, was upregulated in M2 macrophages, and higher FGL2 expression was associated with poorer survival in esophageal carcinoma patients. Using the TIMER database, we found that FGL2 expression correlated positively with the levels of immune markers of infiltrating B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells in esophageal carcinoma samples. Correlation analyses in cBioPortal revealed that the mRNA levels of FGL2 correlated strongly with those of interleukin 10, matrix metalloproteinase 9, C-C motif chemokine ligand 5, T-cell immunoglobulin mucin 3, interleukin 13, vascular cell adhesion molecule 1, macrophage colony-stimulating factor and fibroblast growth factor 7 in esophageal carcinoma tissues. The same cytokines were upregulated when esophageal squamous cell carcinoma cells were co-cultured with M2-like tumor-associated macrophages. Thus, by secreting FGL2, M2-like tumor-associated macrophages may create an immunosuppressive tumor microenvironment that induces the occurrence and progression of esophageal carcinoma.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Ya Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - An Zhi Zhang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Chen Hao Jiang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Fan Ping Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Yu Fang Xie
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Jiang Fen Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Wei Hua Liang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Hai Jun Zhang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Chun Xia Liu
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Li Juan Pang
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Xi Hua Shen
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| | - Feng Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China.,Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jian Ming Hu
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases (Ministry of Education), Department of Pathology, The First Affiliated Hospital, Shihezi University School of Medicine, Xinjiang 832000, China
| |
Collapse
|
41
|
Extracellular ATP: A Feasible Target for Cancer Therapy. Cells 2020; 9:cells9112496. [PMID: 33212982 PMCID: PMC7698494 DOI: 10.3390/cells9112496] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/22/2022] Open
Abstract
Adenosine triphosphate (ATP) is one of the main biochemical components of the tumor microenvironment (TME), where it can promote tumor progression or tumor suppression depending on its concentration and on the specific ecto-nucleotidases and receptors expressed by immune and cancer cells. ATP can be released from cells via both specific and nonspecific pathways. A non-regulated release occurs from dying and damaged cells, whereas active release involves exocytotic granules, plasma membrane-derived microvesicles, specific ATP-binding cassette (ABC) transporters and membrane channels (connexin hemichannels, pannexin 1 (PANX1), calcium homeostasis modulator 1 (CALHM1), volume-regulated anion channels (VRACs) and maxi-anion channels (MACs)). Extracellular ATP acts at P2 purinergic receptors, among which P2X7R is a key mediator of the final ATP-dependent biological effects. Over the years, P2 receptor- or ecto-nucleotidase-targeting for cancer therapy has been proposed and actively investigated, while comparatively fewer studies have explored the suitability of TME ATP as a target. In this review, we briefly summarize the available evidence suggesting that TME ATP has a central role in determining tumor fate and is, therefore, a suitable target for cancer therapy.
Collapse
|
42
|
Zimmermann H. History of ectonucleotidases and their role in purinergic signaling. Biochem Pharmacol 2020; 187:114322. [PMID: 33161020 DOI: 10.1016/j.bcp.2020.114322] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022]
Abstract
Ectonucleotidases are key for purinergic signaling. They control the duration of activity of purinergic receptor agonists. At the same time, they produce hydrolysis products as additional ligands of purinergic receptors. Due to the considerable diversity of enzymes, purinergic receptor ligands and purinergic receptors, deciphering the impact of extracellular purinergic receptor control has become a challenge. The first group of enzymes described were the alkaline phosphatases - at the time not as nucleotide-metabolizing but as nonspecific phosphatases. Enzymes now referred to as nucleoside triphosphate diphosphohydrolases and ecto-5'-nucleotidase were the first and only nucleotide-specific ectonucleotidases identified. And they were the first group of enzymes related to purinergic signaling. Additional research brought to light a surprising number of ectoenzymes with broad substrate specificity, which can also hydrolyze nucleotides. This short overview traces the development of the field and briefly highlights important results and benefits for therapies of human diseases achieved within nearly a century of investigations.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Goethe University, Institute of Cell Biology and Neuroscience, Max-von-Laue-Str. 13, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
43
|
Zeng J, Ning Z, Wang Y, Xiong H. Implications of CD39 in immune-related diseases. Int Immunopharmacol 2020; 89:107055. [PMID: 33045579 DOI: 10.1016/j.intimp.2020.107055] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/10/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
Abstract
Extracellular adenosine triphosphate (eATP) mediates pro-inflammatory responses by recruiting and activating inflammatory cells. CD39 can hydrolyze eATP into adenosine monophosphate (AMP), while CD73 can convert AMP into the immunosuppressive nucleoside adenosine (ADO). CD39 is a rate-limiting enzyme in this cascade, which is regarded as an immunological switch shifting the ATP-mediated pro-inflammatory environment to the ADO- mediated anti-inflammatory status. The CD39 expression can be detected in a wide spectrum of immunocytes, which is under the influence of environmental and genetic factors. It is increasingly suggested that, CD39 participates in some pathophysiological processes, like inflammatory bowel disease (IBD), sepsis, multiple sclerosis (MS), allergic diseases, ischemia-reperfusion (I/R) injury, systemic lupus erythematosus (SLE), diabetes and cancer. Here, we focus on the current understanding of CD39 in immunity, and comprehensively illustrate the diverse CD39 functions within a variety of disorders.
Collapse
Affiliation(s)
- Jianrui Zeng
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Yuzhong Wang
- Department of Neurology and Central Laboratory, Affiliated Hospital of Jining Medical University, Shandong 272000, China.
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China.
| |
Collapse
|
44
|
Chabab G, Barjon C, Bonnefoy N, Lafont V. Pro-tumor γδ T Cells in Human Cancer: Polarization, Mechanisms of Action, and Implications for Therapy. Front Immunol 2020; 11:2186. [PMID: 33042132 PMCID: PMC7524881 DOI: 10.3389/fimmu.2020.02186] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor immune microenvironment contributes to tumor initiation, progression and response to therapy. Among the immune cell subsets that play a role in the tumor microenvironment, innate-like T cells that express T cell receptors composed of γ and δ chains (γδ T cells) are of particular interest. Indeed, γδ T cells contribute to the immune response against many cancers, notably through their powerful effector functions that lead to the elimination of tumor cells and the recruitment of other immune cells. However, their presence in the tumor microenvironment has been associated with poor prognosis in various solid cancers (breast, colon and pancreatic cancer), suggesting that γδ T cells also display pro-tumor activities. In this review, we outline the current evidences of γδ T cell pro-tumor functions in human cancer. We also discuss the factors that favor γδ T cell polarization toward a pro-tumoral phenotype, the characteristics and functions of such cells, and the impact of pro-tumor subsets on γδ T cell-based therapies.
Collapse
Affiliation(s)
- Ghita Chabab
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Clément Barjon
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Nathalie Bonnefoy
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Virginie Lafont
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
45
|
Conversion of extracellular ATP into adenosine: a master switch in renal health and disease. Nat Rev Nephrol 2020; 16:509-524. [PMID: 32641760 DOI: 10.1038/s41581-020-0304-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2020] [Indexed: 12/22/2022]
Abstract
ATP and its ultimate degradation product adenosine are potent extracellular signalling molecules that elicit a variety of pathophysiological functions in the kidney through the activation of P2 and P1 purinergic receptors, respectively. Extracellular purines can modulate immune responses, balancing inflammatory processes and immunosuppression; indeed, alterations in extracellular nucleotide and adenosine signalling determine outcomes of inflammation and healing processes. The functional activities of ectonucleotidases such as CD39 and CD73, which hydrolyse pro-inflammatory ATP to generate immunosuppressive adenosine, are therefore pivotal in acute inflammation. Protracted inflammation may result in aberrant adenosinergic signalling, which serves to sustain inflammasome activation and worsen fibrotic reactions. Alterations in the expression of ectonucleotidases on various immune cells, such as regulatory T cells and macrophages, as well as components of the renal vasculature, control purinergic receptor-mediated effects on target tissues within the kidney. The role of CD39 as a rheostat that can have an impact on purinergic signalling in both acute and chronic inflammation is increasingly supported by the literature, as detailed in this Review. Better understanding of these purinergic processes and development of novel drugs targeting these pathways could lead to effective therapies for the management of acute and chronic kidney disease.
Collapse
|
46
|
Cao W, Fang F, Gould T, Li X, Kim C, Gustafson C, Lambert S, Weyand CM, Goronzy JJ. Ecto-NTPDase CD39 is a negative checkpoint that inhibits follicular helper cell generation. J Clin Invest 2020; 130:3422-3436. [PMID: 32452837 PMCID: PMC7324201 DOI: 10.1172/jci132417] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 03/17/2020] [Indexed: 12/21/2022] Open
Abstract
Vaccination is a mainstay in preventive medicine, reducing morbidity and mortality from infection, largely by generating pathogen-specific neutralizing antibodies. However, standard immunization strategies are insufficient with increasing age due to immunological impediments, including defects in T follicular helper (Tfh) cells. Here, we found that Tfh generation is inversely linked to the expression of the ecto-NTPDase CD39 that modifies purinergic signaling. The lineage-determining transcription factor BCL6 inhibited CD39 expression, while increased Tfh frequencies were found in individuals with a germline polymorphism preventing transcription of ENTPD1, encoding CD39. In in vitro human and in vivo mouse studies, Tfh generation and germinal center responses were enhanced by reducing CD39 expression through the inhibition of the cAMP/PKA/p-CREB pathway, or by blocking adenosine signaling downstream of CD39 using the selective adenosine A2a receptor antagonist istradefylline. Thus, purinergic signaling in differentiating T cells can be targeted to improve vaccine responses, in particular in older individuals who have increased CD39 expression.
Collapse
|
47
|
Repopulation of T, B, and NK cells following alemtuzumab treatment in relapsing-remitting multiple sclerosis. J Neuroinflammation 2020; 17:189. [PMID: 32539719 PMCID: PMC7296935 DOI: 10.1186/s12974-020-01847-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 05/19/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To characterize long-term repopulation of peripheral immune cells following alemtuzumab-induced lymphopenia in relapsing-remitting MS (RRMS), with a focus on regulatory cell types, and to explore associations with clinical outcome measures. METHODS The project was designed as a multicenter add-on longitudinal mechanistic study for RRMS patients enrolled in CARE-MS II, CARE-MS II extension at the University of Southern California and Stanford University, and an investigator-initiated study conducted at the Universities of British Columbia and Chicago. Methods involved collection of blood at baseline, prior to alemtuzumab administration, and at months 5, 11, 17, 23, 36, and 48 post-treatment. T cell, B cell, and natural killer (NK) cell subsets, chemokine receptor expression in T cells, in vitro cytokine secretion patterns, and regulatory T cell (Treg) function were assessed. Clinical outcomes, including expanded disability status score (EDSS), relapses, conventional magnetic resonance imaging (MRI) measures, and incidents of secondary autoimmunity were tracked. RESULTS Variable shifts in lymphocyte populations occurred over time in favor of CD4+ T cells, B cells, and NK cells with surface phenotypes characteristic of regulatory subsets, accompanied by reduced ratios of effector to regulatory cell types. Evidence of increased Treg competence was observed after each treatment course. CD4+ and CD8+ T cells that express CXCR3 and CCR5 and CD8+ T cells that express CDR3 and CCR4 were also enriched after treatment, indicating heightened trafficking potential in activated T cells. Patterns of repopulation were not associated with measures of clinical efficacy or secondary autoimmunity, but exploratory analyses using a random generalized estimating equation (GEE) Poisson model provide preliminary evidence of associations between pro-inflammatory cell types and increased risk for gadolinium (Gd+) enhancing lesions, while regulatory subsets were associated with reduced risk. In addition, the risk for T2 lesions correlated with increases in CD3+CD8+CXCR3+ cells. CONCLUSIONS Lymphocyte repopulation after alemtuzumab treatment favors regulatory subsets in the T cell, B cell, and NK cell compartments. Clinical efficacy may reflect the sum of interactions among them, leading to control of potentially pathogenic effector cell types. Several immune measures were identified as possible biomarkers of lesion activity. Future studies are necessary to more precisely define regulatory and effector subsets and their contributions to clinical efficacy and risk for secondary autoimmunity in alemtuzumab-treated patients, and to reveal new insights into mechanisms of immunopathogenesis in MS. TRIAL REGISTRATION Parent trials for this study are registered with ClinicalTrials.gov: CARE-MS II: NCT00548405, CARE-MS II extension: NCT00930553 and ISS: NCT01307332.
Collapse
|
48
|
Dorneles GP, dos Passos AA, Romão PR, Peres A. New Insights about Regulatory T Cells Distribution and Function with Exercise: The Role of Immunometabolism. Curr Pharm Des 2020; 26:979-990. [DOI: 10.2174/1381612826666200305125210] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/08/2020] [Indexed: 02/08/2023]
Abstract
A lack of physical activity is linked to the development of many chronic diseases through a chronic
low-grade inflammation state. It is now well accepted that the immune system plays a central role in the development
of several chronic diseases, including insulin resistance, type 2 diabetes, atherosclerosis, heart failure and
certain types of cancer. Exercise elicits a strong anti-inflammatory response independently of weight loss and can
be a useful non-pharmacologic strategy to counteract the low-grade inflammation. The CD4+CD25+CD127-
FoxP3+ Regulatory T (Treg) cells are a unique subset of helper T-cells, which regulate immune response and
establish self-tolerance through the secretion of immunoregulatory cytokines, such as IL-10 and TGF-β, and the
suppression of the function and activity of many immune effector cells (including monocytes/macrophages, dendritic
cells, CD4+ and CD8+ T cells, and Natural Killers). The metabolic phenotype of Tregs are regulated by the
transcription factor Foxp3, providing flexibility in fuel choice, but a preference for higher fatty acid oxidation. In
this review, we focus on the mechanisms by which exercise - both acute and chronic - exerts its antiinflammatory
effects through Treg cells mobilization. Furthermore, we discuss the implications of immunometabolic
changes during exercise for the modulation of Treg phenotype and its immunosuppressive function. This
narrative review focuses on the current knowledge regarding the role of Treg cells in the context of acute and
chronic exercise using data from observational and experimental studies. Emerging evidence suggests that the
immunomodulatory effects of exercise are mediated by the ability of exercise to adjust and improve Tregs number
and function.
Collapse
Affiliation(s)
- Gilson P. Dorneles
- Cellular and Molecular Immunology Lab., Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre - RS, Brazil
| | - Aline A.Z. dos Passos
- Cellular and Molecular Immunology Lab., Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre - RS, Brazil
| | - Pedro R.T. Romão
- Cellular and Molecular Immunology Lab., Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre - RS, Brazil
| | - Alessandra Peres
- Cellular and Molecular Immunology Lab., Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre - RS, Brazil
| |
Collapse
|
49
|
Zyma M, Pawliczak R. Characteristics and the role of purinergic receptors in pathophysiology with focus on immune response. Int Rev Immunol 2020; 39:97-117. [PMID: 32037918 DOI: 10.1080/08830185.2020.1723582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The nucleotide adenosine-5'-triphosphate (ATP) is mostly thought to be energy carrier, but evidence presented in multiple studies proves ATP involvement into variety of processes, due to its neuromodulatory capabilities. ATP and its metabolite-adenosine, bind to the purinergic receptors, which are divided into two types: adenosine binding P1 receptor and ADP/ATP binding P2 receptor. These receptors are expressed in different tissues and organs. Recent studies report their immunomodulatory characteristics, connected with varying immunological processes, such as immunological response or antigen presentation. Besides, they seem to play an important role in medical conditions such as bronchial asthma or variety of cancers. In this article, we would like to review recent discoveries on the field of purinergic receptors research focusing on their role in immunological system, and shed a new light upon the importance of these receptors in modern medicine development.
Collapse
Affiliation(s)
- Marharyta Zyma
- Department of Immunopathology, Division of Biomedical Science, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Division of Biomedical Science, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
50
|
Harries M, Hardman J, Chaudhry I, Poblet E, Paus R. Profiling the human hair follicle immune system in lichen planopilaris and frontal fibrosing alopecia: can macrophage polarization differentiate these two conditions microscopically? Br J Dermatol 2020; 183:537-547. [PMID: 31883384 DOI: 10.1111/bjd.18854] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Frontal fibrosing alopecia (FFA) is traditionally regarded as a variant of lichen planopilaris (LPP) based on histological features. Distinct clinical presentation, demographics and epidemiology suggest that differing pathogenic factors determine the final phenotype. OBJECTIVES To map the hair follicle immune system in LPP and FFA by systematically comparing key inflammatory markers in defined hair follicle compartments. METHODS Lesional scalp biopsies from LPP and FFA and healthy controls were stained with the following immunohistochemical markers: CD1a and CD209, CD4, CD8, CD56, CD68, CD123, CXCR3, forkhead box (FOX)P3, mast cell tryptase and cKit. Macrophage polarization was explored using CD206, CD163, CD86, receptor for advanced glycation end products (RAGE), interleukin (IL)-4 and IL-13 on paired lesional and nonlesional LPP and FFA samples. RESULTS Increased numbers of CD8+ , CXCR3+ and FOXP3+ T cells and CD68+ macrophages were identified in the distal hair follicle epithelium and perifollicular mesenchyme in both LPP and FFA compared with controls. In both LPP and FFA, total and degranulated mast cells and CD123+ plasmacytoid dendritic cells were increased in the perifollicular mesenchyme adjacent to the bulge and infundibulum, whereas numbers of CD1a+ and CD209+ dendritic cells were significantly reduced in the infundibulum connective tissue sheath. However, only with CD68 staining was a significant difference between LPP and FFA identified, with greater numbers of CD68+ cells in LPP samples. Furthermore, the identified macrophage polarization markers downregulated CD86 and upregulated CD163 and IL-4 expression in lesional LPP compared with FFA samples. CONCLUSIONS This comparative immunopathological analysis is the first to profile systematically the hair follicle immune system in LPP and FFA. Our analysis highlights a potential role of macrophages in disease pathobiology and suggests that macrophage polarization may differ between LPP and FFA, allowing microscopic differentiation. Linked Comment: Kinoshita-Ise. Br J Dermatol 2020; 183:419-420.
Collapse
Affiliation(s)
- M Harries
- Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre and NIHR Biomedical Research Centre, Manchester, U.K.,Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - J Hardman
- Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - I Chaudhry
- Department of Pathology, Royal Liverpool University Hospital, Liverpool, U.K
| | - E Poblet
- Department of Pathology, University General Hospital of Murcia, Murcia, Spain
| | - R Paus
- Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, U.S.A
| |
Collapse
|