1
|
Zhang Y, Hua J, Chen L. Identifying the plasma metabolome responsible for mediating immune cell action in severe COVID-19: a Mendelian randomization investigation. Front Cell Infect Microbiol 2024; 14:1393432. [PMID: 39224704 PMCID: PMC11366714 DOI: 10.3389/fcimb.2024.1393432] [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: 02/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction The immune response regulates the severity of COVID-19 (sCOVID-19). This study examined the cause-and-effect relationship between immune cell traits (ICTs) and the risk of severe COVID-19. Additionally, we discovered the potential role of plasma metabolome in modulating this risk. Methods Employing data from a genome-wide association study (GWAS), we conducted a two-sample Mendelian randomization (MR) assessment of 731 genetic ICTs and sCOVID-19 (5,101 cases, 1,383,241 controls) incidence. The MR analysis was utilized to further quantitate the degree of plasma metabolome-mediated regulation of immune traits in sCOVID-19. Results The inverse variance weighted method recognized 2 plasma metabolites (PMs) responsible for casual associations between immune cells and sCOVID-19 risk. These included Tridecenedioate (C13:1-DC) which regulated the association between CD27 on IgD- CD38br (OR 0.804, 95% CI 0.699-0.925, p = 0.002) and sCOVID-19 risk (mediated proportion: 18.7%); arginine to citrulline ratio which controlled the relationship of CD39 on monocyte (OR 1.053, 95% CI 1.013-1.094, p = 0.009) with sCOVID-19 risk (mediated proportion: -7.11%). No strong evidence that genetically predicted sCOVID-19 influenced the aforementioned immune traits. Conclusion In this study, we have successfully identified a cause-and-effect relationship between certain ICTs, PMs, and the likelihood of contracting severe COVID-19. Our findings can potentially improve the accuracy of COVID-19 prognostic evaluation and provide valuable insights into the underlying mechanisms of the disease.
Collapse
Affiliation(s)
- Yixia Zhang
- Department of Hematology, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Jie Hua
- Department of Gastroenterology, Jiangsu Province People’s Hospital, Nanjing, China
| | - Liang Chen
- Department of Infectious Diseases, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical College of Nanjing University, Nanjing, China
| |
Collapse
|
2
|
Castiglione Morelli MA, Iuliano A, Viggiani L, Matera I, Pistone A, Schettini SCA, Colucci P, Ostuni A. Redox Balance and Inflammatory Response in Follicular Fluids of Women Recovered by SARS-CoV-2 Infection or Anti-COVID-19 Vaccinated: A Combined Metabolomics and Biochemical Study. Int J Mol Sci 2024; 25:8400. [PMID: 39125969 PMCID: PMC11313332 DOI: 10.3390/ijms25158400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
To date, not many studies have presented evidence of SARS-CoV-2 infecting the female reproductive system. Furthermore, so far, no effect of the administration of anti-COVID 19 vaccines has been reported to affect the quality of oocytes retrieved from women who resorted to assisted reproduction technology (ART). The FF metabolic profiles of women who had been infected by SARS-CoV-2 before IVF treatments or after COVID-19 vaccination were examined by 1H NMR. Immunochemical characterization of proteins and cytokines involved in the redox and inflammatory pathways was performed. The increased expression of SOD2 and NQO1, the lack of alteration of IL-6 and CXCL10 levels, as well as the increased expression of CD39, suggested that, both sharing similar molecular mechanisms or proceeding along different routes, the redox balance is controlled in the FF of both vaccinated and recovered women compared to controls. The lower amount of metabolites known to have proinflammatory activity, i.e., TMAO and lipids, further supported the biochemical results, suggesting that the FF microenvironment is controlled so as to guarantee oocyte quality and does not compromise the outcome of ART. In terms of the number of blastocysts obtained after ICSI and the pregnancy rate, the results are also comforting.
Collapse
Affiliation(s)
| | - Assunta Iuliano
- Center for Reproductive Medicine of “San Carlo” Hospital, 85100 Potenza, Italy; (A.I.); (S.C.A.S.); (P.C.)
| | - Licia Viggiani
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (M.A.C.M.); (L.V.); (I.M.); (A.P.)
| | - Ilenia Matera
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (M.A.C.M.); (L.V.); (I.M.); (A.P.)
| | - Alessandro Pistone
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (M.A.C.M.); (L.V.); (I.M.); (A.P.)
| | - Sergio C. A. Schettini
- Center for Reproductive Medicine of “San Carlo” Hospital, 85100 Potenza, Italy; (A.I.); (S.C.A.S.); (P.C.)
| | - Paola Colucci
- Center for Reproductive Medicine of “San Carlo” Hospital, 85100 Potenza, Italy; (A.I.); (S.C.A.S.); (P.C.)
| | - Angela Ostuni
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (M.A.C.M.); (L.V.); (I.M.); (A.P.)
| |
Collapse
|
3
|
Wu G, Liao J, Zhu X, Zhang Y, Lin Y, Zeng Y, Zhao J, Zhang J, Yao T, Shen X, Li H, Hu L, Zhang W. Shexiang Baoxin Pill enriches Lactobacillus to regulate purine metabolism in patients with stable coronary artery disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155727. [PMID: 38781732 DOI: 10.1016/j.phymed.2024.155727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND It has been clinically confirmed that the Shexiang Baoxin Pill (SBP) dramatically reduces the frequency of angina in patients with stable coronary artery disease (SCAD). However, potential therapeutic mechanism of SBP has not been fully explored. PURPOSE The study explored the therapeutic mechanism of SBP in the treatment of SCAD patients. METHODS We examined the serum metabolic profiles of patients with SCAD following SBP treatment. A rat model of acute myocardial infarction (AMI) was established, and the potential therapeutic mechanism of SBP was explored using metabolomics, transcriptomics, and 16S rRNA sequencing. RESULTS SBP decreased inosine production and improved purine metabolic disorders in patients with SCAD and in animal models of AMI. Inosine was implicated as a potential biomarker for SBP efficacy. Furthermore, SBP inhibited the expression of genes involved in purine metabolism, which are closely associated with thrombosis, inflammation, and platelet function. The regulation of purine metabolism by SBP was associated with the enrichment of Lactobacillus. Finally, the effects of SBP on inosine production and vascular function could be transmitted through the transplantation of fecal microbiota. CONCLUSION Our study reveals a novel mechanism by which SBP regulates purine metabolism by enriching Lactobacillus to exert cardioprotective effects in patients with SCAD. The data also provide previously undocumented evidence indicating that inosine is a potential biomarker for evaluating the efficacy of SBP in the treatment of SCAD.
Collapse
Affiliation(s)
- Gaosong Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jingyu Liao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoyan Zhu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuhao Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan Lin
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuanyuan Zeng
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jing Zhao
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jingfang Zhang
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Tingting Yao
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Xiaoxu Shen
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Liang Hu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Naval Medical University, Shanghai, 200433, China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
4
|
Gambichler T, Rüth J, Goesmann S, Höxtermann S, Skrygan M, Susok L, Becker JC, Overheu O, Schmidt W, Reinacher-Schick A. A Prospective Study Investigating Immune Checkpoint Molecule and CD39 Expression on Peripheral Blood Cells for the Prognostication of COVID-19 Severity and Mortality. Viruses 2024; 16:810. [PMID: 38793691 PMCID: PMC11125582 DOI: 10.3390/v16050810] [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: 05/02/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
In patients with COVID-19, broad panels of immune checkpoint molecules (ICPMs) and the purinergic signaling have not been studied in parallel. We aimed to perform in-depth immunophenotyping of major cell subsets present in human peripheral blood of COVID-19 patients and controls using PD1, TIM3, LAG3, TIGIT, and CD200R, as well as CD39, as markers for the purinergic signaling pathway. We studied 76 COVID-19 patients and 12 healthy controls using peripheral blood mononuclear cells on flow cytometry. Univariable and multivariable statistics were performed. All ICPMs studied were significantly overexpressed on different cell subsets of COVID-19 patients when compared with healthy controls. Elevated lactate dehydrogenase; C-reactive protein; age; and high expression of CD45+, CD39+CD45+, TIM3+CD39+CD4+CD45+, and TIM3+CD39+CD8+CD3+CD4+ cells were significantly associated with severe COVID-19. On multivariable analysis, however, only high expression of CD39+CD45+ (OR 51.4, 95% CI 1.5 to 1763) and TIM3+CD39+CD4+CD3+CD45+ (OR 22.6, 95% CI 1.8 to 277) cells was an independent predictor for severe COVID-19. In conclusion, numerous ICPMs are overexpressed in COVID-19 patients when compared with healthy controls, suggesting a pathophysiological role of these molecules in SARS-CoV-2 infection. However, only TIM3 in co-expression with CD39 remained as a significant independent prognostic ICPM on multivariable analysis. The flow cytometric evaluation of TIM3+CD39+CD4+CD3+CD45+, as well as CD39+CD45+, is a powerful tool for the prognostication of COVID-19 patients on hospital admission.
Collapse
Affiliation(s)
- Thilo Gambichler
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Dermatology, Hospital Dortmund, Faculty of Health/School of Medicine, Witten-Herdecke University, 44137 Dortmund, Germany
- Department of Dermatology, Christian Hospital Unna, 59423 Unna, Germany
| | - Jonas Rüth
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Silke Goesmann
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Stefan Höxtermann
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Marina Skrygan
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Laura Susok
- Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Dermatology, Christian Hospital Unna, 59423 Unna, Germany
| | - Jürgen C. Becker
- Translational Skin Cancer Research, DKTK Partner Site Essen/Düsseldorf, West German Cancer Center, Dermatology, University Duisburg-Essen, 45122 Essen, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Oliver Overheu
- Department for Internal Medicine, Ruhr-University Bochum, 44791 Bochum, Germany
- Department for Hematology and Onoclogy with Palliative Care Unit, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Wolfgang Schmidt
- Department for Internal Medicine, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Anke Reinacher-Schick
- Department for Hematology and Onoclogy with Palliative Care Unit, Ruhr-University Bochum, 44791 Bochum, Germany
| |
Collapse
|
5
|
Díaz-García E, García-Sánchez A, Alfaro E, López-Fernández C, Mañas E, Casitas R, Vega S, Cano-Pumarega I, García-Río F, Cubillos-Zapata C. Dysregulation in CD39/CD73 Axis May Trigger the Upsurge of the Immune Suppressive Agent Adenosine in OSA Patients. Arch Bronconeumol 2024; 60:207-214. [PMID: 38485582 DOI: 10.1016/j.arbres.2024.02.013] [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: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Although higher incidence of cancer represents a major burden for obstructive sleep apnea (OSA) patients, the molecular pathways driving this association are not completely understood. Interestingly, adenosinergic signaling has emerged as a powerful immune checkpoint driving tumor development and progression. METHODS Here, we explored the expression of the adenosinergic ecto-enzymes CD39 and CD73 in T-lymphocytes of OSA patients without any evidence of cancer, as well as their soluble forms in plasma (sCD39 and sCD73), along with adenosine. In addition, we explored the role of intermittent hypoxia (IH) in this context by in vitro models. RESULTS Our results showed that CD39 is upregulated while CD73 is downregulated in OSA T-cells' membrane. Moreover, our findings suggest that IH, through HIF-1, mediates the upregulation of both CD39 and CD73; and that CD73 downregulation could be mediated by a higher release of sCD73 by OSA T-lymphocytes. Importantly, we found that both sCD39 and sCD73 are upregulated in OSA plasma, suggesting T-lymphocytes as a potential source for plasmatic sCD73. Finally, our data propose the alterations in CD39/CD73 axis could underlie the upsurge of adenosine levels in the plasma of OSA patients. CONCLUSION Our study reveals a hypoxia-mediated alteration of the CD39/CD73 axis in OSA patients, which could trigger ADO upregulation, thus potentially contributing to the immune suppressive environment and ultimately facilitating tumor development and progression. Therefore, our data highlights the need for new longitudinal studies evaluating CD39 and/or CD73 as potential cancer-risk prognostic biomarkers in OSA patients.
Collapse
Affiliation(s)
- Elena Díaz-García
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Aldara García-Sánchez
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Servicio de Neumología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Enrique Alfaro
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain; Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
| | - Cristina López-Fernández
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Eva Mañas
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Raquel Casitas
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Sara Vega
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | | | - Francisco García-Río
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain; Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain.
| | - Carolina Cubillos-Zapata
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain.
| |
Collapse
|
6
|
Leão Batista Simões J, Webler Eichler S, Raitz Siqueira ML, de Carvalho Braga G, Bagatini MD. Amyotrophic Lateral Sclerosis in Long-COVID Scenario and the Therapeutic Potential of the Purinergic System in Neuromodulation. Brain Sci 2024; 14:180. [PMID: 38391754 PMCID: PMC10886908 DOI: 10.3390/brainsci14020180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) involves the degeneration of motor neurons and debilitating and possibly fatal symptoms. The COVID-19 pandemic directly affected the quality of life of this group, and the SARS-CoV-2 infection accelerated the present neuroinflammatory process. Furthermore, studies indicate that the infection may have led to the development of the pathology. Thus, the scenario after this pandemic presents "long-lasting COVID" as a disease that affects people who have been infected. From this perspective, studying the pathophysiology behind ALS associated with SARS-CoV-2 infection and possible supporting therapies becomes necessary when we understand the impact on the quality of life of these patients. Thus, the purinergic system was trained to demonstrate how its modulation can add to the treatment, reduce disease progression, and result in better prognoses. From our studies, we highlight the P2X7, P2X4, and A2AR receptors and how their activity can directly influence the ALS pathway.
Collapse
Affiliation(s)
| | | | | | | | - Margarete Dulce Bagatini
- Graduate Program in Medical Sciences, Federal University of Fronteira Sul, Chapecó 89815-899, SC, Brazil
| |
Collapse
|
7
|
Figarella K, Kim J, Ruan W, Mills T, Eltzschig HK, Yuan X. Hypoxia-adenosine axis as therapeutic targets for acute respiratory distress syndrome. Front Immunol 2024; 15:1328565. [PMID: 38312838 PMCID: PMC10835146 DOI: 10.3389/fimmu.2024.1328565] [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: 10/30/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
The human respiratory and circulatory systems collaborate intricately to ensure oxygen delivery to all cells, which is vital for ATP production and maintaining physiological functions and structures. During limited oxygen availability, hypoxia-inducible factors (HIFs) are stabilized and play a fundamental role in maintaining cellular processes for hypoxia adaptation. First discovered during investigations of erythropoietin production regulation, HIFs influence physiological and pathological processes, including development, inflammation, wound healing, and cancer. HIFs promote extracellular adenosine signaling by enhancing adenosine generation and receptor signaling, representing an endogenous feedback mechanism that curbs excessive inflammation, supports injury resolution, and enhances hypoxia tolerance. This is especially important for conditions that involve tissue hypoxia, such as acute respiratory distress syndrome (ARDS), which globally poses significant health challenges without specific treatment options. Consequently, pharmacological strategies to amplify HIF-mediated adenosine production and receptor signaling are of great importance.
Collapse
Affiliation(s)
- Katherine Figarella
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jieun Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Wei Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Holger Klaus Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
8
|
Villacampa A, Alfaro E, Morales C, Díaz-García E, López-Fernández C, Bartha JL, López-Sánchez F, Lorenzo Ó, Moncada S, Sánchez-Ferrer CF, García-Río F, Cubillos-Zapata C, Peiró C. SARS-CoV-2 S protein activates NLRP3 inflammasome and deregulates coagulation factors in endothelial and immune cells. Cell Commun Signal 2024; 22:38. [PMID: 38225643 PMCID: PMC10788971 DOI: 10.1186/s12964-023-01397-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/12/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Hyperinflammation, hypercoagulation and endothelial injury are major findings in acute and post-COVID-19. The SARS-CoV-2 S protein has been detected as an isolated element in human tissues reservoirs and is the main product of mRNA COVID-19 vaccines. We investigated whether the S protein alone triggers pro-inflammatory and pro-coagulant responses in primary cultures of two cell types deeply affected by SARS-CoV-2, such are monocytes and endothelial cells. METHODS In human umbilical vein endothelial cells (HUVEC) and monocytes, the components of NF-κB and the NLRP3 inflammasome system, as well as coagulation regulators, were assessed by qRT-PCR, Western blot, flow cytometry, or indirect immunofluorescence. RESULTS S protein activated NF-κB, promoted pro-inflammatory cytokines release, and triggered the priming and activation of the NLRP3 inflammasome system resulting in mature IL-1β formation in both cell types. This was paralleled by enhanced production of coagulation factors such as von Willebrand factor (vWF), factor VIII or tissue factor, that was mediated, at least in part, by IL-1β. Additionally, S protein failed to enhance ADAMTS-13 levels to counteract the pro-coagulant activity of vWF multimers. Monocytes and HUVEC barely expressed angiotensin-converting enzyme-2. Pharmacological approaches and gene silencing showed that TLR4 receptors mediated the effects of S protein in monocytes, but not in HUVEC. CONCLUSION S protein behaves both as a pro-inflammatory and pro-coagulant stimulus in human monocytes and endothelial cells. Interfering with the receptors or signaling pathways evoked by the S protein may help preventing immune and vascular complications driven by such an isolated viral element. Video Abstract.
Collapse
Affiliation(s)
- Alicia Villacampa
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Enrique Alfaro
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Cristina Morales
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Elena Díaz-García
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Cristina López-Fernández
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - José Luis Bartha
- Department of Obstetrics and Gynecology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Gynecology and Obstetrics Service, La Paz University Hospital, Madrid, Spain
| | | | - Óscar Lorenzo
- Laboratory of Diabetes and Vascular pathology, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Biomedical Research Networking Centre on Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Salvador Moncada
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Vascular Pharmacology and Metabolism (FARMAVASM) group, IdiPAZ, Madrid, Spain
| | - Francisco García-Río
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carolina Cubillos-Zapata
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain.
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain.
| | - Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.
- Vascular Pharmacology and Metabolism (FARMAVASM) group, IdiPAZ, Madrid, Spain.
| |
Collapse
|
9
|
Elsaghir A, El-Sabaa EMW, Zahran AM, Mandour SA, Salama EH, Aboulfotuh S, El-Morshedy RM, Tocci S, Mandour AM, Ali WE, Abdel-Wahid L, Sayed IM, El-Mokhtar MA. Elevated CD39+T-Regulatory Cells and Reduced Levels of Adenosine Indicate a Role for Tolerogenic Signals in the Progression from Moderate to Severe COVID-19. Int J Mol Sci 2023; 24:17614. [PMID: 38139439 PMCID: PMC10744088 DOI: 10.3390/ijms242417614] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Viral infections trigger inflammation by controlling ATP release. CD39 ectoenzymes hydrolyze ATP/ADP to AMP, which is converted by CD73 into anti-inflammatory adenosine (ADO). ADO is an anti-inflammatory and immunosuppressant molecule which can enhance viral persistence and severity. The CD39-CD73-adenosine axis contributes to the immunosuppressive T-reg microenvironment and may affect COVID-19 disease progression. Here, we investigated the link between CD39 expression, mostly on T-regs, and levels of CD73, adenosine, and adenosine receptors with COVID-19 severity and progression. Our study included 73 hospitalized COVID-19 patients, of which 33 were moderately affected and 40 suffered from severe infection. A flow cytometric analysis was used to analyze the frequency of T-regulatory cells (T-regs), CD39+ T-regs, and CD39+CD4+ T-cells. Plasma concentrations of adenosine, IL-10, and TGF-β were quantified via an ELISA. An RT-qPCR was used to analyze the gene expression of CD73 and adenosine receptors (A1, A2A, A2B, and A3). T-reg cells were higher in COVID-19 patients compared to healthy controls (7.4 ± 0.79 vs. 2.4 ± 0.28; p < 0.0001). Patients also had a higher frequency of the CD39+ T-reg subset. In addition, patients who suffered from a severe form of the disease had higher CD39+ T-regs compared with moderately infected patients. CD39+CD4+ T cells were increased in patients compared to the control group. An analysis of serum adenosine levels showed a marked decrease in their levels in patients, particularly those suffering from severe illness. However, this was paralleled with a marked decline in the expression levels of CD73. IL-10 and TGF-β levels were higher in COVID-19; in addition, their values were also higher in the severe group. In conclusion, there are distinct immunological alterations in CD39+ lymphocyte subsets and a dysregulation in the adenosine signaling pathway in COVID-19 patients which may contribute to immune dysfunction and disease progression. Understanding these immunological alterations in the different immune cell subsets and adenosine signaling provides valuable insights into the pathogenesis of the disease and may contribute to the development of novel therapeutic approaches targeting specific immune mechanisms.
Collapse
Affiliation(s)
- Alaa Elsaghir
- Department of Microbiology & Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Ehsan M. W. El-Sabaa
- Department of Microbiology & Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Asmaa M. Zahran
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut 71515, Egypt
| | - Sahar A. Mandour
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, Minia 11566, Egypt
| | - Eman H. Salama
- Department of Clinical Pathology, Faculty of Medicine, Sohag University, Sohag 82524, Egypt
| | - Sahar Aboulfotuh
- Department of Clinical Pathology, Faculty of Medicine, Sohag University, Sohag 82524, Egypt
| | - Reham M. El-Morshedy
- Department of Chest Diseases and Tuberculosis, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Stefania Tocci
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Ahmed Mohamed Mandour
- Department of Anesthesia and ICU, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Wael Esmat Ali
- Department of Clinical Pathology, Faculty of Medicine, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Lobna Abdel-Wahid
- Gastroenterology and Hepatology Unit, Internal Medicine Department, Assiut University, Assiut 71515, Egypt
| | - Ibrahim M. Sayed
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Mohamed A. El-Mokhtar
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos P.O. Box 36, Lebanon
- Department of Medical Microbiology & Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
10
|
Carvalho-Barbosa N, Zeidler JD, Savio LEB, Coutinho-Silva R. Purinergic signaling in the battlefield of viral infections. Purinergic Signal 2023:10.1007/s11302-023-09981-8. [PMID: 38038801 DOI: 10.1007/s11302-023-09981-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
Purinergic signaling has been associated with immune defenses against pathogens such as bacteria, protozoa, fungi, and viruses, acting as a sentinel system that signals to the cells when a threat is present. This review focuses on the roles of purinergic signaling and its therapeutic potential for viral infections. In this context, the purinergic system may play potent antiviral roles by boosting interferon signaling. In other cases, though, it can contribute to a hyperinflammatory response and disease severity, resulting in poor outcomes, such as during flu and potentially COVID-19. Lastly, a third situation may occur since viruses are obligatory intracellular parasites that hijack the host cell machinery for their infection and replication. Viruses such as HIV-1 use the purinergic system to favor their infection and persistence within the host cell. Therefore, understanding the particular nuances of purinergic signaling in each viral infection may contribute to designing proper therapeutic strategies to treat viral diseases.
Collapse
Affiliation(s)
- Nayara Carvalho-Barbosa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Edifício do Centro de Ciências da Saúde, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Julianna Dias Zeidler
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Edifício do Centro de Ciências da Saúde, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Edifício do Centro de Ciências da Saúde, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Robson Coutinho-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Edifício do Centro de Ciências da Saúde, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
11
|
Madera‐Sandoval RL, Cérbulo‐Vázquez A, Arriaga‐Pizano LA, Cabrera‐Rivera GL, Basilio‐Gálvez E, Miranda‐Cruz PE, García de la Rosa MT, Prieto‐Chávez JL, Rivero‐Arredondo SV, Cruz‐Cruz A, Rodríguez‐Hernández D, Salazar‐Ríos ME, Salazar‐Ríos E, Serrano‐Molina ED, De Lira‐Barraza RC, Villanueva‐Compean AH, Esquivel‐Pineda A, Ramírez‐Montes de Oca R, Unzueta‐Marta O, Flores‐Padilla G, Anda‐Garay JC, Sánchez‐Hurtado LA, Calleja‐Alarcón S, Romero‐Gutiérrez L, Torres‐Rosas R, Bonifaz LC, Pelayo R, Márquez‐Márquez E, López‐Macías CIIIR, Ferat‐Osorio E. Potential biomarkers for fatal outcome prognosis in a cohort of hospitalized COVID-19 patients with pre-existing comorbidities. Clin Transl Sci 2023; 16:2687-2699. [PMID: 37873554 PMCID: PMC10719476 DOI: 10.1111/cts.13663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/27/2023] [Accepted: 10/01/2023] [Indexed: 10/25/2023] Open
Abstract
The difficulty in predicting fatal outcomes in patients with coronavirus disease 2019 (COVID-19) impacts the general morbidity and mortality due to severe acute respiratory syndrome-coronavirus 2 infection, as it wears out the hospital services that care for these patients. Unfortunately, in several of the candidates for prognostic biomarkers proposed, the predictive power is compromised when patients have pre-existing comorbidities. A cohort of 147 patients hospitalized for severe COVID-19 was included in a descriptive, observational, single-center, and prospective study. Patients were recruited during the first COVID-19 pandemic wave (April-November 2020). Data were collected from the clinical history whereas immunophenotyping by multiparameter flow cytometry analysis allowed us to assess the expression of surface markers on peripheral leucocyte. Patients were grouped according to the outcome in survivors or non-survivors. The prognostic value of leucocyte, cytokines or HLA-DR, CD39, and CD73 was calculated. Hypertension and chronic renal failure but not obesity and diabetes were conditions more frequent among the deceased patient group. Mixed hypercytokinemia, including inflammatory (IL-6) and anti-inflammatory (IL-10) cytokines, was more evident in deceased patients. In the deceased patient group, lymphopenia with a higher neutrophil-lymphocyte ratio (NLR) value was present. HLA-DR expression and the percentage of CD39+ cells were higher than non-COVID-19 patients but remained similar despite the outcome. Receiver operating characteristic analysis and cutoff value of NLR (69.6%, 9.4), percentage NLR (pNLR; 71.1%, 13.6), and IL-6 (79.7%, 135.2 pg/mL). The expression of HLA-DR, CD39, and CD73, as many serum cytokines (other than IL-6) and chemokines levels do not show prognostic potential, were compared to NLR and pNLR values.
Collapse
Affiliation(s)
- Ruth Lizzeth Madera‐Sandoval
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | | | - Lourdes Andrea Arriaga‐Pizano
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Graciela Libier Cabrera‐Rivera
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
- Posgrado en InmunologíaInstituto Politécnico NacionalCiudad de MéxicoMexico
| | - Edna Basilio‐Gálvez
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
- Posgrado de Ciencias Químicobiológicas, Escuela Nacional de Ciencias BiológicasInstituto Politécnico NacionalCiudad de MéxicoMexico
| | - Patricia Esther Miranda‐Cruz
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - María Teresa García de la Rosa
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
- Posgrado en InmunologíaInstituto Politécnico NacionalCiudad de MéxicoMexico
| | - Jessica Lashkmin Prieto‐Chávez
- Centro de Instrumentos, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Silvia Vanessa Rivero‐Arredondo
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Alonso Cruz‐Cruz
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Daniela Rodríguez‐Hernández
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - María Eugenia Salazar‐Ríos
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Enrique Salazar‐Ríos
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Esli David Serrano‐Molina
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | | | | | - Alejandra Esquivel‐Pineda
- Medicina Interna, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Rubén Ramírez‐Montes de Oca
- Medicina Interna, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Omar Unzueta‐Marta
- Medicina Interna, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Guillermo Flores‐Padilla
- Medicina Interna, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Juan Carlos Anda‐Garay
- Medicina Interna, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Luis Alejandro Sánchez‐Hurtado
- Unidad de Cuidados Intensivos, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Salvador Calleja‐Alarcón
- Unidad de Cuidados Intensivos, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Laura Romero‐Gutiérrez
- Unidad de Cuidados Intensivos, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| | - Rafael Torres‐Rosas
- Laboratorio de Inmunología, Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de OdontologíaUniversidad Autónoma “Benito Juárez” de Oaxaca (UABJO)Oaxaca de JuárezMexico
| | - Laura C. Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
- Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro SocialCiudad de MéxicoMexico
| | - Rosana Pelayo
- Centro de Investigación Biomédica de OrienteIMSSPueblaMexico
- Unidad de Educación e Investigación, IMSSCiudad de MéxicoMexico
| | | | | | - Eduardo Ferat‐Osorio
- Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro SocialCiudad de MéxicoMexico
- División de Investigación en Salud, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXIInstituto Mexicano del Seguro Social (IMSS)Ciudad de MéxicoMexico
| |
Collapse
|
12
|
Liu Y, Li Z, Zhao X, Xiao J, Bi J, Li XY, Chen G, Lu L. Review immune response of targeting CD39 in cancer. Biomark Res 2023; 11:63. [PMID: 37287049 DOI: 10.1186/s40364-023-00500-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/11/2023] [Indexed: 06/09/2023] Open
Abstract
The ATP-adenosine pathway has emerged as a promising target for cancer therapy, but challenges remain in achieving effective tumor control. Early research focused on blocking the adenosine generating enzyme CD73 and the adenosine receptors A2AR or A2BR in cancer. However, recent studies have shown that targeting CD39, the rate-limiting ecto-enzyme of the ATP-adenosine pathway, can provide more profound anti-tumor efficacy by reducing immune-suppressive adenosine accumulation and increasing pro-inflammatory ATP levels. In addition, combining CD39 blocking antibody with PD-1 immune checkpoint therapy may have synergistic anti-tumor effects and improve patient survival. This review will discuss the immune components that respond to CD39 targeting in the tumor microenvironment. Targeting CD39 in cancer has been shown to not only decrease adenosine levels in the tumor microenvironment (TME), but also increase ATP levels. Additionally, targeting CD39 can limit the function of Treg cells, which are known to express high levels of CD39. With phase I clinical trials of CD39 targeting currently underway, further understanding and rational design of this approach for cancer therapy are expected.
Collapse
Affiliation(s)
- Yao Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China
| | - Zhongliang Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China
| | - Xiaoguang Zhao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China
| | - Jing Xiao
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jiacheng Bi
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xian-Yang Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China.
- Department of R&D, OriCell Therapeutics Co. Ltd, No.1227, Zhangheng Rd, Pudong, Shanghai, China.
| | - Guokai Chen
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, Guangdong, P.R. China.
| |
Collapse
|
13
|
Arruvito L, Sananez I, Seery V, Russo C, Geffner J. Purinergic signaling pathway in severe COVID-19. Curr Opin Pharmacol 2023; 70:102379. [PMID: 37087844 PMCID: PMC10091870 DOI: 10.1016/j.coph.2023.102379] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 02/19/2023] [Accepted: 03/16/2023] [Indexed: 04/05/2023]
Abstract
Substantial efforts have been made to understand the immune response during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, in order to identify and characterize risk factors, immune mechanisms responsible for the induction of tissue injury and potential therapeutic targets. Purinergic signaling pathway has shown to modulate the inflammatory processes in the course of several infectious diseases, but its role in the coronavirus disease 2019 (COVID-19) has not been clearly defined. Inflammation is usually associated to the release of ATP from different cell types, starting a cascade of events through the activation of a set of different purinergic receptors. This review summarizes the evidence showing the involvement of the purinergic system in the inflammatory condition that characterizes severe COVID-19.
Collapse
Affiliation(s)
- Lourdes Arruvito
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina. UBA-CONICET, Paraguay 2155, C1121ABG CABA, Argentina.
| | - Inés Sananez
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina. UBA-CONICET, Paraguay 2155, C1121ABG CABA, Argentina
| | - Vanesa Seery
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina. UBA-CONICET, Paraguay 2155, C1121ABG CABA, Argentina
| | - Constanza Russo
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina. UBA-CONICET, Paraguay 2155, C1121ABG CABA, Argentina
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina. UBA-CONICET, Paraguay 2155, C1121ABG CABA, Argentina
| |
Collapse
|
14
|
Korb VG, Schultz IC, Beckenkamp LR, Wink MR. A Systematic Review of the Role of Purinergic Signalling Pathway in the Treatment of COVID-19. Int J Mol Sci 2023; 24:ijms24097865. [PMID: 37175571 PMCID: PMC10178215 DOI: 10.3390/ijms24097865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/27/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global health concern. Three years since its origin, despite the approval of vaccines and specific treatments against this new coronavirus, there are still high rates of infection, hospitalization, and mortality in some countries. COVID-19 is characterised by a high inflammatory state and coagulation disturbances that may be linked to purinergic signalling molecules such as adenosine triphosphate (ATP), adenosine diphosphate (ADP), adenosine (ADO), and purinergic receptors (P1 and P2). These nucleotides/nucleosides play important roles in cellular processes, such as immunomodulation, blood clot formation, and vasodilation, which are affected during SARS-CoV-2 infection. Therefore, drugs targeting this purinergic pathway, currently used for other pathologies, are being evaluated in preclinical and clinical trials for COVID-19. In this review, we focus on the potential of these drugs to control the release, degradation, and reuptake of these extracellular nucleotides and nucleosides to treat COVID-19. Drugs targeting the P1 receptors could have therapeutic efficacy due to their capacity to modulate the cytokine storm and the immune response. Those acting in P2X7, which is linked to NLRP3 inflammasome activation, are also valuable candidates as they can reduce the release of pro-inflammatory cytokines. However, according to the available preclinical and clinical data, the most promising medications to be used for COVID-19 treatment are those that modulate platelets behaviour and blood coagulation factors, mainly through the P2Y12 receptor.
Collapse
Affiliation(s)
- Vitoria Guero Korb
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil
| | - Iago Carvalho Schultz
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil
| | - Liziane Raquel Beckenkamp
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil
| | - Márcia Rosângela Wink
- Laboratório de Biologia Celular, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS, Brazil
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Sala 304 Centro, Porto Alegre 90050-170, RS, Brazil
| |
Collapse
|
15
|
Ming LG, Hu DX, Zuo C, Zhang WJ. G protein-coupled P2Y12 receptor is involved in the progression of neuropathic pain. Biomed Pharmacother 2023; 162:114713. [PMID: 37084563 DOI: 10.1016/j.biopha.2023.114713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023] Open
Abstract
The pathological mechanism of neuropathic pain is complex, which seriously affects the physical and mental health of patients, and its treatment is also difficult. The role of G protein-coupled P2Y12 receptor in pain has been widely recognized and affirmed. After nerve injury, stimulated cells can release large amounts of nucleotides into the extracellular matrix, act on P2Y12 receptor. Activated P2Y12 receptor activates intracellular signal transduction and is involved in the development of pain. P2Y12 receptor activation can sensitize primary sensory neurons and receive sensory information. By transmitting the integrated information through the dorsal root of the spinal cord to the secondary neurons of the posterior horn of the spinal cord. The integrated information is then transmitted to the higher center through the ascending conduction tract to produce pain. Moreover, activation of P2Y12 receptor can mediate immune cells to release pro-inflammatory factors, increase damage to nerve cells, and aggravate pain. While inhibits the activation of P2Y12 receptor can effectively relieve pain. Therefore, in this article, we described P2Y12 receptor antagonists and their pharmacological properties. In addition, we explored the potential link between P2Y12 receptor and the nervous system, discussed the intrinsic link of P2Y12 receptor and neuropathic pain and as a potential pharmacological target for pain suppression.
Collapse
Affiliation(s)
- Li-Guo Ming
- Department of Gastrointestinal surgery, the Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, the Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Cheng Zuo
- Department of Gastrointestinal surgery, the Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, the Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China.
| |
Collapse
|
16
|
Elsaghir A, El-Sabaa EMW, Ahmed AK, Abdelwahab SF, Sayed IM, El-Mokhtar MA. The Role of Cluster of Differentiation 39 (CD39) and Purinergic Signaling Pathway in Viral Infections. Pathogens 2023; 12:279. [PMID: 36839551 PMCID: PMC9967413 DOI: 10.3390/pathogens12020279] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
CD39 is a marker of immune cells such as lymphocytes and monocytes. The CD39/CD73 pathway hydrolyzes ATP into adenosine, which has a potent immunosuppressive effect. CD39 regulates the function of a variety of immunologic cells through the purinergic signaling pathways. CD39+ T cells have been implicated in viral infections, including Human Immunodeficiency Virus (HIV), Cytomegalovirus (CMV), viral hepatitis, and Corona Virus Disease 2019 (COVID-19) infections. The expression of CD39 is an indicator of lymphocyte exhaustion, which develops during chronicity. During RNA viral infections, the CD39 marker can profile the populations of CD4+ T lymphocytes into two populations, T-effector lymphocytes, and T-regulatory lymphocytes, where CD39 is predominantly expressed on the T-regulatory cells. The level of CD39 in T lymphocytes can predict the disease progression, antiviral immune responses, and the response to antiviral drugs. Besides, the percentage of CD39 and CD73 in B lymphocytes and monocytes can affect the status of viral infections. In this review, we investigate the impact of CD39 and CD39-expressing cells on viral infections and how the frequency and percentage of CD39+ immunologic cells determine disease prognosis.
Collapse
Affiliation(s)
- Alaa Elsaghir
- Department of Microbiology & Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Ehsan M. W. El-Sabaa
- Department of Microbiology & Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | | | - Sayed F. Abdelwahab
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ibrahim M. Sayed
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Mohamed A. El-Mokhtar
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
17
|
Rud J, Riker RR, Eldridge A, Lord C, deKay JT, May TL, Gagnon DJ, Sawyer D, Ryzhov S, Seder DB. Decreased circulating CD73 and adenosine deaminase are associated with disease severity in hospitalized patients with COVID-19. Int J Immunopathol Pharmacol 2023; 37:3946320231185703. [PMID: 37364162 PMCID: PMC10300631 DOI: 10.1177/03946320231185703] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
OBJECTIVE SARS-CoV-2 infection has been shown to result in increased circulating levels of adenosine triphosphate and adenosine diphosphate and decreased levels of adenosine, which has important anti-inflammatory activity. The goal of this pilot project was to assess the levels of soluble CD73 and soluble Adenosine Deaminase (ADA) in hospitalized patients with COVID-19 and determine if levels of these molecules are associated with disease severity. METHODS Plasma from 28 PCR-confirmed hospitalized COVID-19 patients who had varied disease severity based on WHO classification (6 mild/moderate, 10 severe, 12 critical) had concentrations of both soluble CD73 and ADA determined by ELISA. These concentrations were compared to healthy control plasma that is commercially available and was biobanked prior to the start of the pandemic. Additionally, outcomes such as WHO ordinal scale for disease severity, ICU admission, needed for invasive ventilation, hospital length of stay, and development of thrombosis during admission were used as markers of disease severity. RESULTS Our results show that both CD73 and ADA are decreased during SARS-CoV-2 infection. The level of circulating CD73 is directly correlated to the severity of the disease defined by the need for ICU admission, invasive ventilation, and hospital length of stay. Low level of CD73 is also associated with clinical thrombosis, a severe complication of SARS-CoV-2 infection. CONCLUSION Our study indicates that adenosine metabolism is down-regulated in patients with COVID-19 and associated with severe infection. Further large-scale studies are warranted to investigate the role of the adenosinergic anti-inflammatory CD73/ADA axis in protection against COVID-19.
Collapse
Affiliation(s)
- Jonathan Rud
- Department of Acute Care/Hospital Medicine, Maine General Medical Center, Augusta, ME, USA
- Maine Health Institute for Research, Scarborough, ME, USA
| | - Richard R Riker
- Maine Health Institute for Research, Scarborough, ME, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Ashley Eldridge
- Maine Health Institute for Research, Scarborough, ME, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Christine Lord
- Maine Health Institute for Research, Scarborough, ME, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Joanne T deKay
- Maine Health Institute for Research, Scarborough, ME, USA
| | - Teresa L May
- Maine Health Institute for Research, Scarborough, ME, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - David J Gagnon
- Maine Health Institute for Research, Scarborough, ME, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Douglas Sawyer
- Maine Health Institute for Research, Scarborough, ME, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| | - Sergey Ryzhov
- Maine Health Institute for Research, Scarborough, ME, USA
| | - David B Seder
- Maine Health Institute for Research, Scarborough, ME, USA
- Department of Critical Care Services, Maine Medical Center, Portland, ME, USA
| |
Collapse
|
18
|
Bencze D, Fekete T, Pfliegler W, Szöőr Á, Csoma E, Szántó A, Tarr T, Bácsi A, Kemény L, Veréb Z, Pázmándi K. Interactions between the NLRP3-Dependent IL-1β and the Type I Interferon Pathways in Human Plasmacytoid Dendritic Cells. Int J Mol Sci 2022; 23:ijms232012154. [PMID: 36293012 PMCID: PMC9602791 DOI: 10.3390/ijms232012154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Generally, a reciprocal antagonistic interaction exists between the antiviral type I interferon (IFN) and the antibacterial nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3)-dependent IL-1β pathways that can significantly shape immune responses. Plasmacytoid dendritic cells (pDCs), as professional type I IFN-producing cells, are the major coordinators of antiviral immunity; however, their NLRP3-dependent IL-1β secretory pathway is poorly studied. Our aim was to determine the functional activity of the IL-1β pathway and its possible interaction with the type I IFN pathway in pDCs. We found that potent nuclear factor-kappa B (NF-κB) inducers promote higher levels of pro-IL-1β during priming compared to those activation signals, which mainly trigger interferon regulatory factor (IRF)-mediated type I IFN production. The generation of cleaved IL-1β requires certain secondary signals in pDCs and IFN-α or type I IFN-inducing viruses inhibit IL-1β production of pDCs, presumably by promoting the expression of various NLRP3 pathway inhibitors. In line with that, we detected significantly lower IL-1β production in pDCs of psoriasis patients with elevated IFN-α levels. Collectively, our results show that the NLRP3-dependent IL-1β secretory pathway is inducible in pDCs; however, it may only prevail under inflammatory conditions, in which the type I IFN pathway is not dominant.
Collapse
Affiliation(s)
- Dóra Bencze
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Walter Pfliegler
- Department of Molecular Biotechnology and Microbiology, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Eszter Csoma
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Antónia Szántó
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tünde Tarr
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Lajos Kemény
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Kitti Pázmándi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: ; Tel./Fax: +36-52-417-159
| |
Collapse
|
19
|
Pietrobon AJ, Andrejew R, Custódio RWA, Oliveira LDM, Scholl JN, Teixeira FME, de Brito CA, Glaser T, Kazmierski J, Goffinet C, Turdo AC, Yendo T, Aoki V, Figueiró F, Battastini AM, Ulrich H, Benard G, Duarte AJDS, Sato MN. Dysfunctional purinergic signaling correlates with disease severity in COVID-19 patients. Front Immunol 2022; 13:1012027. [PMID: 36248842 PMCID: PMC9562777 DOI: 10.3389/fimmu.2022.1012027] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022] Open
Abstract
Ectonucleotidases modulate inflammatory responses by balancing extracellular ATP and adenosine (ADO) and might be involved in COVID-19 immunopathogenesis. Here, we explored the contribution of extracellular nucleotide metabolism to COVID-19 severity in mild and severe cases of the disease. We verified that the gene expression of ectonucleotidases is reduced in the whole blood of patients with COVID-19 and is negatively correlated to levels of CRP, an inflammatory marker of disease severity. In line with these findings, COVID-19 patients present higher ATP levels in plasma and reduced levels of ADO when compared to healthy controls. Cell type-specific analysis revealed higher frequencies of CD39+ T cells in severely ill patients, while CD4+ and CD8+ expressing CD73 are reduced in this same group. The frequency of B cells CD39+CD73+ is also decreased during acute COVID-19. Interestingly, B cells from COVID-19 patients showed a reduced capacity to hydrolyze ATP into ADP and ADO. Furthermore, impaired expression of ADO receptors and a compromised activation of its signaling pathway is observed in COVID-19 patients. The presence of ADO in vitro, however, suppressed inflammatory responses triggered in patients’ cells. In summary, our findings support the idea that alterations in the metabolism of extracellular purines contribute to immune dysregulation during COVID-19, possibly favoring disease severity, and suggest that ADO may be a therapeutic approach for the disease.
Collapse
Affiliation(s)
- Anna Julia Pietrobon
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Roberta Andrejew
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Ricardo Wesley Alberca Custódio
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Luana de Mendonça Oliveira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Juliete Nathali Scholl
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Franciane Mouradian Emidio Teixeira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cyro Alves de Brito
- Technical Division of Medical Biology, Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Julia Kazmierski
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department and Division of Infectious and Parasitic Diseases, Berlin Institute of Health, Berlin, Germany
| | - Christine Goffinet
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department and Division of Infectious and Parasitic Diseases, Berlin Institute of Health, Berlin, Germany
| | - Anna Claudia Turdo
- Department and Division of Infectious and Parasitic Diseases, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Tatiana Yendo
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Valeria Aoki
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Fabricio Figueiró
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana Maria Battastini
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Gill Benard
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Alberto Jose da Silva Duarte
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Maria Notomi Sato
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
- *Correspondence: Maria Notomi Sato,
| |
Collapse
|
20
|
Abstract
On 4 September, 2020, the US National Institutes of Health launched a new clinical trial, “A Multicenter, Adaptive, Randomized Controlled Platform Trial of the Safety and Efficacy of Antithrombotic and Additional Strategies in Hospitalized Adults with COVID-19.” This open-label, placebo-controlled, multicenter, adaptive platform study was designed to evaluate therapeutic options for patients hospitalized with mild, moderate, or severe COVID-19. A variety of drugs and drug classes were selected, including heparin, the monoclonal antibody crizanlizumab, sodium-glucose cotransporter-2 inhibitors, and purinergic signaling receptor Y12 inhibitors. These medications have been widely used in the treatment of other conditions, from sick cell disease to type 2 diabetes mellitus and some forms of cardiovascular disease, but their inclusion in a study of COVID-19 was somewhat unexpected. This article examines the rationale behind the use of these disparate agents in the treatment and prevention of adverse outcomes in patients with COVID-19 and explores how these strategies may be utilized in the future to address the severe acute respiratory syndrome coronavirus 2 pandemic.
Collapse
Affiliation(s)
- Matthew W McCarthy
- Department of Medicine, Weill Cornell Medicine, 525 East 68th Street, Box 130, New York, NY, 10065, USA.
| |
Collapse
|
21
|
Wienkamp AK, Erpenbeck L, Rossaint J. Platelets in the NETworks interweaving inflammation and thrombosis. Front Immunol 2022; 13:953129. [PMID: 35979369 PMCID: PMC9376363 DOI: 10.3389/fimmu.2022.953129] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/07/2022] [Indexed: 12/18/2022] Open
Abstract
Platelets are well characterized for their indispensable role in primary hemostasis to control hemorrhage. Research over the past years has provided a substantial body of evidence demonstrating that platelets also participate in host innate immunity. The surface expression of pattern recognition receptors, such as TLR2 and TLR4, provides platelets with the ability to sense bacterial products in their environment. Platelet α-granules contain microbicidal proteins, chemokines and growth factors, which upon release may directly engage pathogens and/or contribute to inflammatory signaling. Additionally, platelet interactions with neutrophils enhance neutrophil activation and are often crucial to induce a sufficient immune response. In particular, platelets can activate neutrophils to form neutrophil extracellular traps (NETs). This specific neutrophil effector function is characterized by neutrophils expelling chromatin fibres decorated with histones and antimicrobial proteins into the extracellular space where they serve to trap and kill pathogens. Until now, the mechanisms and signaling pathways between platelets and neutrophils inducing NET formation are still not fully characterized. NETs were also detected in thrombotic lesions in several disease backgrounds, pointing towards a role as an interface between neutrophils, platelets and thrombosis, also known as immunothrombosis. The negatively charged DNA within NETs provides a procoagulant surface, and in particular NET-derived proteins may directly activate platelets. In light of the current COVID-19 pandemic, the topic of immunothrombosis has become more relevant than ever, as a majority of COVID-19 patients display thrombi in the lung capillaries and other vascular beds. Furthermore, NETs can be found in the lung and other tissues and are associated with an increased mortality. Here, virus infiltration may lead to a cytokine storm that potently activates neutrophils and leads to massive neutrophil infiltration into the lung and NET formation. The resulting NETs presumably activate platelets and coagulation factors, further contributing to the subsequent emergence of microthrombi in pulmonary capillaries. In this review, we will discuss the interplay between platelets and NETs and the potential of this alliance to influence the course of inflammatory diseases. A better understanding of the underlying molecular mechanisms and the identification of treatment targets is of utmost importance to increase patients’ survival and improve the clinical outcome.
Collapse
Affiliation(s)
- Ann-Katrin Wienkamp
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Luise Erpenbeck
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
- *Correspondence: Jan Rossaint,
| |
Collapse
|
22
|
Alfaro E, Díaz-García E, García-Tovar S, Zamarrón E, Mangas A, Galera R, Nanwani-Nanwani K, Pérez-de-Diego R, López-Collazo E, García-Río F, Cubillos-Zapata C. Impaired Kallikrein-Kinin System in COVID-19 Patients' Severity. Front Immunol 2022; 13:909342. [PMID: 35812405 PMCID: PMC9258198 DOI: 10.3389/fimmu.2022.909342] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/12/2022] [Indexed: 12/27/2022] Open
Abstract
COVID-19 has emerged as a devastating disease in the last 2 years. Many authors appointed to the importance of kallikrein-kinin system (KKS) in COVID-19 pathophysiology as it is involved in inflammation, vascular homeostasis, and coagulation. We aim to study the bradykinin cascade and its involvement in severity of patients with COVID-19. This is an observational cohort study involving 63 consecutive patients with severe COVID-19 pneumonia and 27 healthy subjects as control group. Clinical laboratory findings and plasma protein concentration of KKS peptides [bradykinin (BK), BK1-8], KKS proteins [high–molecular weight kininogen (HK)], and KKS enzymes [carboxypeptidase N subunit 1 (CPN1), kallikrein B1 (KLKB1), angiotensin converting enzyme 2 (ACE2), and C1 esterase inhibitor (C1INH)] were analyzed. We detected dysregulated KKS in patients with COVID-19, characterized by an accumulation of BK1-8 in combination with decreased levels of BK. Accumulated BK1-8 was related to severity of patients with COVID-19. A multivariate logistic regression model retained BK1-8, BK, and D-dimer as independent predictor factors to intensive care unit (ICU) admission. A Youden’s optimal cutoff value of −0.352 was found for the multivariate model score with an accuracy of 92.9%. Multivariate model score-high group presented an odds ratio for ICU admission of 260.0. BK1-8 was related to inflammation, coagulation, and lymphopenia. Our data suggest that BK1-8/BK plasma concentration in combination with D-dimer levels might be retained as independent predictors for ICU admission in patients with COVID-19. Moreover, we reported KKS dysregulation in patients with COVID-19, which was related to disease severity by means of inflammation, hypercoagulation, and lymphopenia.
Collapse
Affiliation(s)
- Enrique Alfaro
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Elena Díaz-García
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Sara García-Tovar
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Ester Zamarrón
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Alberto Mangas
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Raúl Galera
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | | | - Rebeca Pérez-de-Diego
- Laboratory of Immunogenetics of Human Diseases, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | | | - Francisco García-Río
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
- *Correspondence: Francisco García-Río, ; Carolina Cubillos-Zapata,
| | - Carolina Cubillos-Zapata
- Respiratory Diseases Group, Respiratory Service, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
- *Correspondence: Francisco García-Río, ; Carolina Cubillos-Zapata,
| |
Collapse
|