1
|
Cruz-Gregorio A, Amezcua-Guerra LM, Fisher-Bautista B, Romero-Beltrán A, Fonseca-Camarillo G. The Protective Role of Interleukin-37 in Cardiovascular Diseases through Ferroptosis Modulation. Int J Mol Sci 2024; 25:9758. [PMID: 39337246 PMCID: PMC11432013 DOI: 10.3390/ijms25189758] [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: 07/29/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
The role of ferroptosis and iron metabolism dysregulation in the pathophysiology of cardiovascular diseases is increasingly recognized. Conditions such as hypertension, cardiomyopathy, atherosclerosis, myocardial ischemia/reperfusion injury, heart failure, and cardiovascular complications associated with COVID-19 have been linked to these processes. Inflammation is central to these conditions, prompting exploration into the inflammatory and immunoregulatory molecular pathways that mediate ferroptosis and its contribution to cardiovascular disease progression. Notably, emerging evidence highlights interleukin-37 as a protective cytokine with the ability to activate the nuclear factor erythroid 2-related factor 2 pathway, inhibit macrophage ferroptosis, and attenuate atherosclerosis progression in murine models. However, a comprehensive review focusing on interleukin-37 and its protective role against ferroptosis in CVD is currently lacking. This review aims to fill this gap by summarizing existing knowledge on interleukin-37, including its regulatory functions and impact on ferroptosis in conditions such as atherosclerosis and myocardial infarction. We also explore experimental strategies and propose that targeting interleukin-37 to modulate ferroptosis presents a promising therapeutic approach for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Alfredo Cruz-Gregorio
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Luis M Amezcua-Guerra
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
- Departamento de Atención a la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 14387, Mexico
| | - Brandon Fisher-Bautista
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
- Programa de Maestría en Ciencias Químico Biológicas, Instituto Politécnico Nacional, Ciudad de México 11350, Mexico
| | - Abraham Romero-Beltrán
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
- Departamento de Atención a la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México 14387, Mexico
| | - Gabriela Fonseca-Camarillo
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| |
Collapse
|
2
|
Sanna FC, Benešová I, Pervan P, Krenz A, Wurzel A, Lohmayer R, Mühlbauer J, Wöllner A, Köhl N, Menevse AN, Stamova S, Volpin V, Beckhove P, Xydia M. IL-2 and TCR stimulation induce expression and secretion of IL-32β by human T cells. Front Immunol 2024; 15:1437224. [PMID: 39211051 PMCID: PMC11357969 DOI: 10.3389/fimmu.2024.1437224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
IL-32 expression is important for pathogen clearance but detrimental in chronic inflammation, autoimmunity, and cancer. T cells are major IL-32 producers in these diseases and key mediators of pathogen and tumor elimination but also autoimmune destruction. However, their contribution to IL-32 biology during immune responses is hardly understood due to several isoforms with divergent inflammatory properties. Here, we identified IL-32β as the predominant isoform in various T cell subsets of healthy individuals and breast cancer patients with the highest levels detected in intratumoral regulatory T cells. We show that IL-32β is induced by IL-2 but IL-32β release requires T Cell Receptor rather than IL2R stimulation. Using inhibitors of protein secretion pathways and serial (ultra)centrifugation of T cell supernatants, we demonstrate that T cells actively secrete IL-32β unconventionally, as a free protein and, to a minor degree, through exosomes. Thus, our data identify activated T cells as major IL-32β secretors in health and cancer.
Collapse
Affiliation(s)
| | - Iva Benešová
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Philip Pervan
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Adriana Krenz
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Alexander Wurzel
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Robert Lohmayer
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
- Algorithmic Bioinformatics, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Jasmin Mühlbauer
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Amélie Wöllner
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Nina Köhl
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| | - Ayse Nur Menevse
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Slava Stamova
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Valentina Volpin
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Philipp Beckhove
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Medical Center, Regensburg, Germany
| | - Maria Xydia
- Interventional Immunology, Leibniz Institute for Immunotherapy, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), Regensburg, Germany
| |
Collapse
|
3
|
Kumar V, Narisawa M, Cheng XW. Overview of multifunctional Tregs in cardiovascular disease: From insights into cellular functions to clinical implications. FASEB J 2024; 38:e23786. [PMID: 38979903 DOI: 10.1096/fj.202400839r] [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: 04/12/2024] [Revised: 06/01/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
Regulatory T cells (Tregs) are crucial in regulating T-cell-mediated immune responses. Numerous studies have shown that dysfunction or decreased numbers of Tregs may be involved in inflammatory cardiovascular diseases (CVDs) such as atherosclerosis, hypertension, myocardial infarction, myocarditis, cardiomyopathy, valvular heart diseases, heart failure, and abdominal aortic aneurysm. Tregs can help to ameliorate CVDs by suppressing excessive inflammation through various mechanisms, including inhibition of T cells and B cells, inhibition of macrophage-induced inflammation, inhibition of dendritic cells and foam cell formation, and induction of anti-inflammatory macrophages. Enhancing or restoring the immunosuppressive activity of Tregs may thus serve as a fundamental immunotherapy to treat hypertension and CVDs. However, the precise molecular mechanisms underlying the Tregs-induced protection against hypertension and CVDs remain to be investigated. This review focuses on recent advances in our understanding of Tregs subsets and function in CVDs. In addition, we discuss promising strategies for using Tregs through various pharmacological approaches to treat hypertension and CVDs.
Collapse
Affiliation(s)
- Vipin Kumar
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P.R. China
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P.R. China
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
4
|
Shang J, Xu Y, Pu S, Sun X, Gao X. Role of IL-34 and its receptors in inflammatory diseases. Cytokine 2023; 171:156348. [PMID: 37683444 DOI: 10.1016/j.cyto.2023.156348] [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: 08/06/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023]
Abstract
In recent years, IL-34 has been widely discussed as a novel cytokine. IL-34 is a pro-inflammatory cytokine binding four distinct receptors, namely CSF-1R, syndecan-1, PTP-ζ and TREM2. Previous studies have shown that IL-34 and its receptors play important roles in the development and progression of various inflammatory diseases. Therefore, IL-34 has the potential to be a biomarker and therapeutic target for inflammatory diseases. However, further study is still needed to identify the specific mechanism through which IL-34 contributes to illness. In this article, we review the recent advances in the biological roles of IL-34 and its receptors as well as their roles in the development and therapeutic application of inflammatory diseases.
Collapse
Affiliation(s)
- Jiameng Shang
- The First Affiliated Hospital of Harbin Medical University, People's Republic of China
| | - Yuxin Xu
- The First Affiliated Hospital of Harbin Medical University, People's Republic of China
| | - Shengdan Pu
- The First Affiliated Hospital of Harbin Medical University, People's Republic of China
| | - Xiaotong Sun
- The First Affiliated Hospital of Harbin Medical University, People's Republic of China
| | - Xinyuan Gao
- The First Affiliated Hospital of Harbin Medical University, People's Republic of China.
| |
Collapse
|
5
|
Gu M, Jin Y, Gao X, Xia W, Xu T, Pan S. Novel insights into IL-37: an anti-inflammatory cytokine with emerging roles in anti-cancer process. Front Immunol 2023; 14:1278521. [PMID: 37928545 PMCID: PMC10623001 DOI: 10.3389/fimmu.2023.1278521] [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: 08/16/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Interleukin-37 (IL-37) is a newly discovered member of IL-1 family. The cytokine was proved to have extensive protective effects in infectious diseases, allergic diseases, metabolic diseases, autoimmune diseases and tumors since its discovery. IL-37 was mainly produced by immune and some non-immune cells in response to inflammatory stimulus. The IL-37 precursors can convert into the mature forms after caspase-1 cleavage and activation intracellularly, and then bind to Smad-3 and transfer to the nucleus to inhibit the production and functions of proinflammatory cytokines; extracellularly, IL-37 binds to cell surface receptors to form IL-37/IL-18Rα/IL-1R8 complex to exert immunosuppressive function via inhibiting/activating multiple signal pathways. In addition, IL-37 can attenuate the pro-inflammatory effect of IL-18 through directly or forming an IL-37/IL-18BP/IL-18Rβ complex. Therefore, IL-37 has the ability to suppress innate and acquired immunity of the host, and effectively control inflammatory stimulation, which was considered as a new hallmark of cancer. Specifically, it is concluded that IL-37 can inhibit the growth and migration of tumor cells, prohibit angiogenesis and mediate the immunoregulation in tumor microenvironment, so as to exert effective anti-tumor effects. Importantly, latest studies also showed that IL-37 may be a novel therapeutic target for cancer monitoring. In this review, we summarize the immunoregulation roles and mechanisms of IL-37 in anti-tumor process, and discuss its progress so far and potential as tumor immunotherapy.
Collapse
Affiliation(s)
- Min Gu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Yuexinzi Jin
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Xun Gao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Wenying Xia
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Ting Xu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Shiyang Pan
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| |
Collapse
|
6
|
Lu C, Zhou F, Xian H, Sun S, Yue J, Zhang Y, Zhao Q, Luo X, Li Y. Serum IL-38 Level Was Associated with Incidence of MACE in the STEMI Patients. Int J Gen Med 2023; 16:2987-2997. [PMID: 37465556 PMCID: PMC10350401 DOI: 10.2147/ijgm.s417471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023] Open
Abstract
Background The relationship between serum IL-38 and major adverse cardiovascular events (MACE) in patients with ST elevation myocardial infarction (STEMI) remains unclear. Methods In the present study, 589 STEMI patients were included, the serum level of IL-38 was measured. The median follow-up time was 720 days, the STEMI patients were divided into high IL-38 (IL-38>6.49ng/mL) and low IL-38 groups (IL-38≤6.49ng/mL) to compare the probability of MACE. Results Plasma IL-38 levels were significantly lower in STEMI patients than in SAP patients (4.0±2.2 vs 6.9±3.2 ng/mL, P < 0.001). Ninety-three STEMI patients met the defined MACE study endpoint. The incidence of MACE was significantly lower in patients with high IL-38 group than in patients with low IL-38 group (7.8% vs 23.7%, P < 0.001). Low plasma IL-38 levels were independently associated with the occurrence of MACE (OR = 0.90, P < 0.001). Conclusion We get a conclusion that low plasma levels of IL-38 are independently associated with the occurrence of MACE.
Collapse
Affiliation(s)
- Chengbo Lu
- Department of Cardiology, 1st Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, People’s Republic of China
| | - Fanghui Zhou
- Department of Hematology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| | - Huimin Xian
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| | - Siyuan Sun
- Department of Cardiology, People’s Hospital of Taihe County, Taihe, Anhui, People’s Republic of China
| | - Jingkun Yue
- Department of Cardiology, People’s Hospital of Taihe County, Taihe, Anhui, People’s Republic of China
| | - Ying Zhang
- Department of Gynaecology and Obstetrics, 1st Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, People’s Republic of China
| | - Qi Zhao
- Department of Cardiology, 1st Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| | - Xing Luo
- Department of Cardiology, 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| | - Yang Li
- Department of Cardiology, 4th Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
7
|
Xu J, Han X, Xia N, Zhao Q, Cheng Z. IL‑37 suppresses macrophage ferroptosis to attenuate diabetic atherosclerosis via the NRF2 pathway. Exp Ther Med 2023; 25:289. [PMID: 37206550 PMCID: PMC10189585 DOI: 10.3892/etm.2023.11988] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/16/2023] [Indexed: 05/21/2023] Open
Abstract
IL-37 is a newly discovered inflammatory factor. However, the protective effect and underlying mechanisms of IL-37 on atherosclerosis remain unclear. In the present study, IL-37 was used for intraperitoneal injection in diabetic ApoE-/- mice caused by streptozotocin. High glucose (HG)/ox-LDL was used to stimulate THP-1 original macrophage followed by IL-37 pretreatment in vitro. The atheromatous plaque area, oxidative stress and inflammation levels in ApoE-/- mice were evaluated, and the level of macrophage ferroptosis was detected in vivo and in vitro. It was identified that IL-37 treatment significantly decreased plaque area in diabetic ApoE-/- mice. IL-37 not only improved blood lipid levels in mice, but also reduced serum levels of inflammatory factors including IL-1β and IL-18. Furthermore, IL-37 increased GPX4 and nuclear factor erythroid 2-related factor 2 (NRF2) in the aorta of diabetic mice. In vitro experiment revealed that IL-37 inhibited HG/ox-LDL-induced ferroptosis in macrophages, as evidenced by improved cell membrane oxidation, reduced malondialdehyde production and increased GPX4 expression. Moreover, it was also found that IL-37 enhanced the nuclear translocation of NRF2 in macrophages, while ML385, a specific NRF2 inhibitor, significantly attenuated the protective effect of IL-37 on macrophage ferroptosis caused by HG/ox-LDL. In conclusion, IL-37 suppressed macrophage ferroptosis to attenuate atherosclerosis progression via activating the NRF2 pathway.
Collapse
Affiliation(s)
- Jinmei Xu
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xu Han
- Medical Laboratory, Harbin Medical University, Daqing, Heilongjiang 163001, P.R. China
| | - Nan Xia
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Qingsong Zhao
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Zhifeng Cheng
- Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
- Correspondence to: Dr Zhifeng Cheng, Department of Endocrinology, Fourth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
8
|
Tomasi M, Cherubini A, Pelusi S, Margarita S, Bianco C, Malvestiti F, Miano L, Romeo S, Prati D, Valenti L. Circulating Interlukin-32 and Altered Blood Pressure Control in Individuals with Metabolic Dysfunction. Int J Mol Sci 2023; 24:ijms24087465. [PMID: 37108628 PMCID: PMC10138906 DOI: 10.3390/ijms24087465] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Fatty liver disease is most frequently related to metabolic dysfunction (MAFLD) and associated comorbidities, heightening the risk of cardiovascular disease, and is associated with higher hepatic production of IL32, a cytokine linked with lipotoxicity and endothelial activation. The aim of this study was to examine the relationship between circulating IL32 concentration and blood pressure control in individuals with metabolic dysfunction at high risk of MAFLD. IL32 plasma levels were measured by ELISA in 948 individuals with metabolic dysfunction enrolled in the Liver-Bible-2021 cohort. Higher circulating IL32 levels were independently associated with systolic blood pressure (estimate +0.008 log10 per 1 mmHg increase, 95% c.i. 0.002-0.015; p = 0.016), and inversely correlated with antihypertensive medications (estimate -0.189, 95% c.i. -0.291--0.088, p = 0.0002). Through multivariable analysis, IL32 levels predicted both systolic blood pressure (estimate 0.746, 95% c.i 0.173-1.318; p = 0.010) and impaired blood pressure control (OR 1.22, 95% c.i. 1.09-1.38; p = 0.0009) independently of demographic and metabolic confounders and of treatment. This study reveals that circulating IL32 levels are associated with impaired blood pressure control in individuals at risk of cardiovascular disease.
Collapse
Affiliation(s)
- Melissa Tomasi
- Precision Medicine Lab-Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Alessandro Cherubini
- Precision Medicine Lab-Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Serena Pelusi
- Precision Medicine Lab-Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Sara Margarita
- Precision Medicine Lab-Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Cristiana Bianco
- Precision Medicine Lab-Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesco Malvestiti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Lorenzo Miano
- Precision Medicine Lab-Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, 413 45 Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Science, University Magna Graecia, 88100 Catanzaro, Italy
| | - Daniele Prati
- Precision Medicine Lab-Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Luca Valenti
- Precision Medicine Lab-Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
9
|
Khattar G, Hallit J, El Chamieh C, Bou Sanayeh E. Wellens' syndrome following severe COVID-19 infection, an innocent coincidence or a deadly association: two case reports. BMC Cardiovasc Disord 2023; 23:106. [PMID: 36829118 PMCID: PMC9950701 DOI: 10.1186/s12872-023-03137-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 02/20/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has been associated with late-onset cardiovascular complications primarily due to a hypercoagulable state. Its association with Wellens' syndrome, which reflects a stenosis in the proximal left anterior descending coronary artery, is not well established. We present two cases diagnosed with this syndrome following their COVID-19 acute phase despite taking adequate anticoagulation. CASE PRESENTATION We present two patients with incidental electrocardiography (ECG) showing the typical Wellens'-related changes, with an underlying severe triple-vessel coronary artery disease a few weeks following a severe COVID-19 infection associated with high inflammatory markers. The stenotic lesions were diagnosed by cardiac catheterization, and both patients underwent Coronary Artery Bypass Grafting successfully. Notably, patients' baseline ECGs were normal, and they were maintained on Rivaroxaban 10 mg following their viral illness. CONCLUSION Despite advances in the preventive measures for COVID-19 complications, its pathophysiologic impact on vasculature and atherosclerosis is still incompletely understood. Further clinical trials must be conducted to study this association between Wellens' syndrome and this virus to prevent life-threatening complications.
Collapse
Affiliation(s)
- Georges Khattar
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon.,Notre Dame des Secours University Hospital, Jbeil, Lebanon
| | - Jennifer Hallit
- Department of Pediatrics, American University of Beirut Medical Center, Beirut, Lebanon
| | - Carolla El Chamieh
- Faculty of Medicine and Medical Sciences, Holy Spirit University of Kaslik (USEK), Jounieh, Lebanon
| | - Elie Bou Sanayeh
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
10
|
Makarova YA, Ryabkova VA, Salukhov VV, Sagun BV, Korovin AE, Churilov LP. Atherosclerosis, Cardiovascular Disorders and COVID-19: Comorbid Pathogenesis. Diagnostics (Basel) 2023; 13:478. [PMID: 36766583 PMCID: PMC9914751 DOI: 10.3390/diagnostics13030478] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
The article describes how atherosclerosis and coronavirus disease 19 (COVID-19) may affect each other. The features of this comorbid pathogenesis at various levels (vascular, cellular and molecular) are considered. A bidirectional influence of these conditions is described: the presence of cardiovascular diseases affects different individuals' susceptibility to viral infection. In turn, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can have a negative effect on the endothelium and cardiomyocytes, causing blood clotting, secretion of pro-inflammatory cytokines, and thus exacerbating the development of atherosclerosis. In addition to the established entry into cells via angiotensin-converting enzyme 2 (ACE2), other mechanisms of SARS-CoV-2 entry are currently under investigation, for example, through CD147. Pathogenesis of comorbidity can be determined by the influence of the virus on various links which are meaningful for atherogenesis: generation of oxidized forms of low-density lipoproteins (LDL), launch of a cytokine storm, damage to the endothelial glycocalyx, and mitochondrial injury. The transformation of a stable plaque into an unstable one plays an important role in the pathogenesis of atherosclerosis complications and can be triggered by COVID-19. The impact of SARS-CoV-2 on large vessels such as the aorta is more complex than previously thought considering its impact on vasa vasorum. Current information on the mutual influence of the medicines used in the treatment of atherosclerosis and acute COVID-19 is briefly summarized.
Collapse
Affiliation(s)
- Yulia A. Makarova
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Saint Petersburg State University, 199034 Saint-Petersburg, Russia
| | - Varvara A. Ryabkova
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Saint Petersburg State University, 199034 Saint-Petersburg, Russia
- M.V. Chernorutsky Department of Internal Medicine (Hospital Course), Pavlov First Saint Petersburg State Medical University, 197022 Saint-Petersburg, Russia
| | - Vladimir V. Salukhov
- N.S. Molchanov 1st Clinic for the Improvement of Physicians, S.M. Kirov Military Medical Academy, 194044 Saint-Petersburg, Russia
| | - Boris V. Sagun
- N.S. Molchanov 1st Clinic for the Improvement of Physicians, S.M. Kirov Military Medical Academy, 194044 Saint-Petersburg, Russia
| | - Aleksandr E. Korovin
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Saint Petersburg State University, 199034 Saint-Petersburg, Russia
- N.S. Molchanov 1st Clinic for the Improvement of Physicians, S.M. Kirov Military Medical Academy, 194044 Saint-Petersburg, Russia
| | - Leonid P. Churilov
- Laboratory of the Microangiopathic Mechanisms of Atherogenesis, Saint Petersburg State University, 199034 Saint-Petersburg, Russia
- Department of Experimental Tuberculosis, Saint Petersburg Research Institute of Phthisiopulmonology, 191036 Saint-Petersburg, Russia
| |
Collapse
|
11
|
Kaufmann CC, Ahmed A, Muthspiel M, Rostocki I, Pogran E, Zweiker D, Burger AL, Jäger B, Aicher G, Spiel AO, Vafai-Tabrizi F, Gschwantler M, Fasching P, Wojta J, Huber K. Association of Interleukin-32 and Interleukin-34 with Cardiovascular Disease and Short-Term Mortality in COVID-19. J Clin Med 2023; 12:jcm12030975. [PMID: 36769623 PMCID: PMC9917403 DOI: 10.3390/jcm12030975] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Excess cardiovascular (CV) morbidity and mortality has been observed in patients with COVID-19. Both interleukin-32 (IL-32) and interleukin-34 (IL-34) have been hypothesized to contribute to CV involvement in COVID-19. METHODS This prospective, observational study of patients with laboratory-confirmed COVID-19 infection was conducted from 6 June to 22 December 2020 in a tertiary care hospital in Vienna, Austria. IL-32 and IL-34 levels on admission were collected and tested for their association with CV disease and short-term mortality in patients with COVID-19. CV disease was defined by the presence of coronary artery disease, heart failure, stroke or atrial fibrillation and patients were stratified by CV disease burden. RESULTS A total of 245 eligible patients with COVID-19 were included, of whom 37 (15.1%) reached the primary endpoint of 28-day mortality. Of the total sample, 161 had no CV disease (65.7%), 69 had one or two CV diseases (28.2%) and 15 patients had ≥three CV diseases (6.1%). Median levels of IL-32 and IL-34 at admission were comparable across the three groups of CV disease burden. IL-32 and IL-34 failed to predict mortality upon both univariable and multivariable Cox regression analysis. The two CV disease groups, however, had a significantly higher risk of mortality within 28 days (one or two CV diseases: crude HR 4.085 (95% CI, 1.913-8.725), p < 0.001 and ≥three CV diseases: crude HR 13.173 (95% CI, 5.425-31.985), p < 0.001). This association persisted for those with ≥three CV diseases after adjustment for age, gender and CV risk factors (adjusted HR 3.942 (95% CI, 1.288-12.068), p = 0.016). CONCLUSION In our study population of hospitalized patients with COVID-19, IL-32 and IL-34 did not show any associations with CV disease or 28-day mortality in the context of COVID-19. Patients with multiple CV diseases, however, had a significantly increased risk of short-term mortality.
Collapse
Affiliation(s)
- Christoph C. Kaufmann
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
- Correspondence: ; Tel.: +43-1-49150-2301; Fax: +43-1-49150-2309
| | - Amro Ahmed
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Marie Muthspiel
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Isabella Rostocki
- Department of Endocrinology and Rheumatology, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Edita Pogran
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - David Zweiker
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
- Division of Cardiology, Medical University of Graz, 8036 Graz, Austria
| | - Achim Leo Burger
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Bernhard Jäger
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Gabriele Aicher
- Department of Laboratory Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Alexander O. Spiel
- Department of Emergency Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Florian Vafai-Tabrizi
- 2nd Medical Department with Pneumology and Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Michael Gschwantler
- Department of Gastroenterology and Hepatology, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
- Medical School, Sigmund Freud University, 1020 Vienna, Austria
| | - Peter Fasching
- Department of Endocrinology and Rheumatology, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine 2, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria
- Core Facilities, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for CV Research, 1090 Vienna, Austria
| | - Kurt Huber
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), 1160 Vienna, Austria
- Medical School, Sigmund Freud University, 1020 Vienna, Austria
- Ludwig Boltzmann Institute for CV Research, 1090 Vienna, Austria
| |
Collapse
|
12
|
Xiang C, Li H, Tang W. Targeting CSF-1R represents an effective strategy in modulating inflammatory diseases. Pharmacol Res 2023; 187:106566. [PMID: 36423789 DOI: 10.1016/j.phrs.2022.106566] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Colony-stimulating factor-1 receptor (CSF-1R), also known as FMS kinase, is a type I single transmembrane protein mainly expressed in myeloid cells, such as monocytes, macrophages, glial cells, and osteoclasts. The endogenous ligands, colony-stimulating factor-1 (CSF-1) and Interleukin-34 (IL-34), activate CSF-1R and downstream signaling pathways including PI3K-AKT, JAK-STATs, and MAPKs, and modulate the proliferation, differentiation, migration, and activation of target immune cells. Over the past decades, the promising therapeutic potential of CSF-1R signaling inhibition has been widely studied for decreasing immune suppression and escape in tumors, owing to depletion and reprogramming of tumor-associated macrophages. In addition, the excessive activation of CSF-1R in inflammatory diseases is consecutively uncovered in recent years, which may result in inflammation in bone, kidney, lung, liver and central nervous system. Agents against CSF-1R signaling have been increasingly investigated in preclinical or clinical studies for inflammatory diseases treatment. However, the pathological mechanism of CSF-1R in inflammation is indistinct and whether CSF-1R signaling can be identified as biomarkers remains controversial. With the background information aforementioned, this review focus on the dialectical roles of CSF-1R and its ligands in regulating innate immune cells and highlights various therapeutic implications of blocking CSF-1R signaling in inflammatory diseases.
Collapse
Affiliation(s)
- Caigui Xiang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heng Li
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Wei Tang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
13
|
Zamani B, Najafizadeh M, Motedayyen H, Arefnezhad R. Predicting roles of IL-27 and IL-32 in determining the severity and outcome of COVID-19. Int J Immunopathol Pharmacol 2022; 36:3946320221145827. [PMID: 36476070 PMCID: PMC9742516 DOI: 10.1177/03946320221145827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Immune changes play fundamental roles in the pathogenesis and severity of coronavirus disease 2019 (COVID-19). Previous studies have revealed alterations in immune responses of patients with non-severe and severe COVID-19. Therefore, this study investigated whether interleukin-27 (IL-27) and interleukin-32 (IL-32) levels may be considered as predicting factors for determining the severity and outcome of COVID-19. METHODS The blood samples were collected from 50 non-severe and severe patients infected with COVID-19 and 25 healthy subjects. The serum samples were isolated from the whole blood. The levels of IL-27 and IL-32 were measured by enzyme-linked immunosorbent assay and percentages of some immune cells were studied by cell counter. RESULTS The levels of IL-27 and IL-32 were significantly higher in COVID-19 patients than healthy subjects (p < 0.0001-0.01). IL-27 was significantly reduced in severe COVID-19 patients who needed to undergo ICU therapy (p < 0.05). Disease severity was significantly associated with IL-27 level in patients with COVID-19 (p < 0.05), unlike IL-32 level. There was a significant association between IL-27 and IL-32 in participants (p < 0.0001, odds ratio (OR) = 0.9873; 95% confidence interval (CI) = 0.9998 to 1.000; p < 0.05, OR = 0.4462; 95% CI = 0.08,579 to 0.7802; p < 0.01, OR = 0.6640, 95% CI = 0.3007-0.8590). IL-27 level was significantly higher in the recovered subjects than dead cases (p < 0.0001). IL-27 and IL-32 levels in patients who had fever were significantly higher than those who did not have (p < 0.01-0.05), unlike patients who suffered from cough (p < 0.001-0.01). The IL-27 level in patients with non-severe COVID-19 was directly correlated with CRP value (p < 0.05, OR = 0.5,722,357, 95% CI = 0.06,807,176-0.8,435,928). IL-27 and IL-32 levels in non-severe patients were positively associated with NLR (p < 0.01, OR = 0.7292; 95% CI = 0.2809 to 0.9163; p < 0.01, OR = 0.6537, 95% CI = 0.1425-0.8896). Patients with severe COVID-19 had a significant increase in NLR (p < 0.0001-0.05). NLR was significantly correlated with the disease severity (p < 0.0001-0.05). Survivors had a significant reduction in NLR compared with those who succumbed to COVID-19 (p < 0.05). CONCLUSION Change in IL-27 level along with the frequencies of some immune cells may serve as a predictor of the severity and outcome of COVID-19.
Collapse
Affiliation(s)
- Batool Zamani
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Maedeh Najafizadeh
- Infectious Disease Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran,Hossein Motedayyen, Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, 5th Kilometer of Ravand Road, Kashan, Iran. ; Reza ArefNezhad, Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran. reza.aref1374@gmail
| | - Reza Arefnezhad
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
14
|
Ma Y, Su H, Wang X, Niu X, Che Y, Hambly BD, Bao S, Wang X. The role of IL-35 and IL-37 in breast cancer - potential therapeutic targets for precision medicine. Front Oncol 2022; 12:1051282. [PMID: 36483045 PMCID: PMC9723453 DOI: 10.3389/fonc.2022.1051282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2023] Open
Abstract
Breast cancer is still a major concern due to its relatively poor prognosis in women, although there are many approaches being developed for the management of breast cancer. Extensive studies demonstrate that the development of breast cancer is determined by pro versus anti tumorigenesis factors, which are closely related to host immunity. IL-35 and IL-37, anti-inflammatory cytokines, play an important role in the maintenance of immune homeostasis. The current review focuses on the correlation between clinical presentations and the expression of IL-35 and IL-37, as well as the potential underlying mechanism during the development of breast cancer in vitro and in vivo. IL-35 is inversely correlated the differentiation and prognosis in breast cancer patients; whereas IL-37 shows dual roles during the development of breast cancer, and may be breast cancer stage dependent. Such information might be useful for both basic scientists and medical practitioners in the management of breast cancer patients.
Collapse
Affiliation(s)
- Yuntao Ma
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - He Su
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xuyun Wang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiangdong Niu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yang Che
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Brett D Hambly
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, Australia
| | - Shisan Bao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xiaopeng Wang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
15
|
Sex Differences in Coronary Artery Disease and Diabetes Revealed by scRNA-Seq and CITE-Seq of Human CD4+ T Cells. Int J Mol Sci 2022; 23:ijms23179875. [PMID: 36077273 PMCID: PMC9456056 DOI: 10.3390/ijms23179875] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022] Open
Abstract
Despite the decades-old knowledge that males and people with diabetes mellitus (DM) are at increased risk for coronary artery disease (CAD), the reasons for this association are only partially understood. Among the immune cells involved, recent evidence supports a critical role of T cells as drivers and modifiers of CAD. CD4+ T cells are commonly found in atherosclerotic plaques. We aimed to understand the relationship of CAD with sex and DM by single-cell RNA (scRNA-Seq) and antibody sequencing (CITE-Seq) of CD4+ T cells. Peripheral blood mononuclear cells (PBMCs) of 61 men and women who underwent cardiac catheterization were interrogated by scRNA-Seq combined with 49 surface markers (CITE-Seq). CAD severity was quantified using Gensini scores, with scores above 30 considered CAD+ and below 6 considered CAD-. Four pairs of groups were matched for clinical and demographic parameters. To test how sex and DM changed cell proportions and gene expression, we compared matched groups of men and women, as well as diabetic and non-diabetic subjects. We analyzed 41,782 single CD4+ T cell transcriptomes for sex differences in 16 women and 45 men with and without coronary artery disease and with and without DM. We identified 16 clusters in CD4+ T cells. The proportion of cells in CD4+ effector memory cluster 8 (CD4T8, CCR2+ Em) was significantly decreased in CAD+, especially among DM+ participants. This same cluster, CD4T8, was significantly decreased in female participants, along with two other CD4+ T cell clusters. In CD4+ T cells, 31 genes showed significant and coordinated upregulation in both CAD and DM. The DM gene signature was partially additive to the CAD gene signature. We conclude that (1) CAD and DM are clearly reflected in PBMC transcriptomes, and (2) significant differences exist between women and men and (3) between subjects with DM and non-DM.
Collapse
|
16
|
Dang J, He Z, Cui X, Fan J, Hambly DJ, Hambly BD, Li X, Bao S. The Role of IL-37 and IL-38 in Colorectal Cancer. Front Med (Lausanne) 2022; 9:811025. [PMID: 35186997 PMCID: PMC8847758 DOI: 10.3389/fmed.2022.811025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/10/2022] [Indexed: 12/25/2022] Open
Abstract
Colorectal cancer (CRC) is a major killer. Dysregulation of IL-37 and IL-38, both anti-inflammatory cytokines, is observed in auto-immune diseases. The precise regulatory mechanisms of IL-37/IL-38 during the development of CRC remains unclear, but chronic intestinal inflammation is involved in the carcinogenesis of CRC. Constitutive production of colonic IL-37 and IL-38 is substantially reduced in CRC, consistent with an inverse correlation with CRC differentiation. Reduced colonic IL-37 and IL-38 is relating to CRC invasion and distant metastasis, suggesting a protective role for IL-38 within the tumor micro-environment. IL-38 is reduced in right-sided CRC compared to left-sided CRC, which is in line with multiple risk factors for right-sided CRC, including the embryonic development of the colon, and genetic differences in CRC between these two sides. Finally, colonic IL-37 and tumor associated neutrophils (TAN) seem to be independent biomarkers of prognostic value, whereas colonic IL-38 seems to be a reliable and independent biomarker in predicting the 5-year survival post-surgery in CRC. However, there is room for improvement in available studies, including the extension of these studies to different regions/countries incorporating different races, evaluation of the role of multi-drug resistance, and different subsets of CRC. It would be useful to determine the kinetics of circulating IL-38 and its relationship with drug resistance/targeted therapy. The measurement of colonic IL-38 at the molecular and cellular level is required to explore the contribution of IL-38 pathways during the development of CRC. These approaches could provide insight for the development of personalized medicine.
Collapse
Affiliation(s)
- Jie Dang
- Child and Adolescent Health Management Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhiyun He
- Department of General Surgery, Lanzhou University First Hospital, Lanzhou, China
| | - Xiang Cui
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Jingchun Fan
- Department of Epidemiology and Evidence-Based Medicine, School of Public Health, Centre for Evidence-Based Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - David J Hambly
- Resident Training Program, Gold Coast University Hospital, Southport, QLD, Australia
| | - Brett D Hambly
- Department of Epidemiology and Evidence-Based Medicine, School of Public Health, Centre for Evidence-Based Medicine, Gansu University of Chinese Medicine, Lanzhou, China.,Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, Australia
| | - Xun Li
- Department of General Surgery, Lanzhou University First Hospital, Lanzhou, China
| | - Shisan Bao
- Department of Epidemiology and Evidence-Based Medicine, School of Public Health, Centre for Evidence-Based Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
17
|
Tokajian S, Merhi G, Al Khoury C, Nemer G. Interleukin-37: A Link Between COVID-19, Diabetes, and the Black Fungus. Front Microbiol 2022; 12:788741. [PMID: 35095801 PMCID: PMC8793130 DOI: 10.3389/fmicb.2021.788741] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/23/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic involved millions of people and diabetes was identified as an associated comorbidity. Initiation of systemic corticosteroids in patients suffering from severe COVID-19 was associated with lower mortality. A surge of invasive fungal infections of the maxillofacial region, namely mucormycosis, was linked to a deadly infection known as black fungus. Black fungus, diabetes, corticosteroids, and coronavirus disease 2019 (COVID-19) all have a dysregulated immune response in common, which partly could also be attributed to interleukin 37 (IL-37). IL-37, a new cytokine of the IL-1 family, known for broadly reducing innate inflammation as well as acquired immune responses. The use of corticosteroids in diabetic COVID-19 patients, crowded hospitals, and lack of medical oxygen should be carefully considered to reduce COVID-associated secondary infections.
Collapse
Affiliation(s)
- Sima Tokajian
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos, Lebanon,*Correspondence: Sima Tokajian,
| | - Georgi Merhi
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos, Lebanon
| | - Charbel Al Khoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos, Lebanon
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon,Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
18
|
Sun X, Gao J, Meng X, Lu X, Zhang L, Chen R. Polarized Macrophages in Periodontitis: Characteristics, Function, and Molecular Signaling. Front Immunol 2021; 12:763334. [PMID: 34950140 PMCID: PMC8688840 DOI: 10.3389/fimmu.2021.763334] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2021] [Indexed: 12/23/2022] Open
Abstract
Periodontitis (PD) is a common chronic infectious disease. The local inflammatory response in the host may cause the destruction of supporting periodontal tissue. Macrophages play a variety of roles in PD, including regulatory and phagocytosis. Moreover, under the induction of different factors, macrophages polarize and form different functional phenotypes. Among them, M1-type macrophages with proinflammatory functions and M2-type macrophages with anti-inflammatory functions are the most representative, and both of them can regulate the tendency of the immune system to exert proinflammatory or anti-inflammatory functions. M1 and M2 macrophages are involved in the destructive and reparative stages of PD. Due to the complex microenvironment of PD, the dynamic development of PD, and various local mediators, increasing attention has been given to the study of macrophage polarization in PD. This review summarizes the role of macrophage polarization in the development of PD and its research progress.
Collapse
Affiliation(s)
- Xiaoyu Sun
- *Correspondence: Lei Zhang, ; Xiaoyu Sun,
| | | | | | | | - Lei Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, Department of Periodontology, Stomatologic Hospital & College, Anhui Medical University, Hefei, China
| | | |
Collapse
|