1
|
Lee JH, Shin SJ, Lee JH, Knowles JC, Lee HH, Kim HW. Adaptive immunity of materials: Implications for tissue healing and regeneration. Bioact Mater 2024; 41:499-522. [PMID: 39206299 PMCID: PMC11350271 DOI: 10.1016/j.bioactmat.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024] Open
Abstract
Recent cumulative findings signify the adaptive immunity of materials as a key agenda in tissue healing that can improve regenerative events and outcomes. Modulating immune responses, mainly the recruitment and functions of T and B cells and their further interplay with innate immune cells (e.g., dendritic cells, macrophages) can be orchestrated by materials. For instance, decellularized matrices have been shown to promote muscle healing by inducing T helper 2 (Th2) cell immunity, while synthetic biopolymers exhibit differential effects on B cell responses and fibrosis compared decellularized matrices. We discuss the recent findings on how implantable materials instruct the adaptive immune events and the subsequent tissue healing process. In particular, we dissect the materials' physicochemical properties (shape, size, topology, degradation, rigidity, and matrix dynamic mechanics) to demonstrate the relations of these parameters with the adaptive immune responses in vitro and the underlying biological mechanisms. Furthermore, we present evidence of recent in vivo phenomena, including tissue healing, cancer progression, and fibrosis, wherein biomaterials potentially shape adaptive immune cell functions and in vivo outcomes. Our discussion will help understand the materials-regulated immunology events more deeply, and offer the design rationale of materials with tunable matrix properties for accelerated tissue repair and regeneration.
Collapse
Affiliation(s)
- Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jin Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jonathan C. Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman Dental Institute, University College London, London NW3 2PX, United Kingdom
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
2
|
Li J, Yi X, Liu L, Wang X, Ai J. Advances in tumor nanotechnology: theragnostic implications in tumors via targeting regulated cell death. Apoptosis 2023:10.1007/s10495-023-01851-3. [PMID: 37184582 DOI: 10.1007/s10495-023-01851-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2023] [Indexed: 05/16/2023]
Abstract
Cell death constitutes an indispensable part of the organismal balance in the human body. Generally, cell death includes regulated cell death (RCD) and accidental cell death (ACD), reflecting the intricately molecule-dependent process and the uncontrolled response, respectively. Furthermore, diverse RCD pathways correlate with multiple diseases, such as tumors and neurodegenerative diseases. Meanwhile, with the development of precision medicine, novel nano-based materials have gradually been applied in the clinical diagnosis and treatment of tumor patients. As the carrier, organic, inorganic, and biomimetic nanomaterials could facilitate the distribution, improve solubility and bioavailability, enhance biocompatibility and decrease the toxicity of drugs in the body, therefore, benefiting tumor patients with better survival outcomes and quality of life. In terms of the most studied cell death pathways, such as apoptosis, necroptosis, and pyroptosis, plenty of studies have explored specific types of nanomaterials targeting the molecules and signals in these pathways. However, no attempt was made to display diverse nanomaterials targeting different RCD pathways comprehensively. In this review, we elaborate on the potential mechanisms of RCD, including intrinsic and extrinsic apoptosis, necroptosis, ferroptosis, pyroptosis, autophagy-dependent cell death, and other cell death pathways together with corresponding nanomaterials. The thorough presentation of RCD pathways and diverse nano-based materials may provide a wider cellular and molecular landscape of tumor diagnosis and treatments.
Collapse
Affiliation(s)
- Jin Li
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xianyanling Yi
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Liangren Liu
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.
| | - Jianzhong Ai
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
High-Dose Intravenous Ascorbate in Sepsis, a Pro-Oxidant Enhanced Microbicidal Activity and the Effect on Neutrophil Functions. Biomedicines 2022; 11:biomedicines11010051. [PMID: 36672559 PMCID: PMC9855518 DOI: 10.3390/biomedicines11010051] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Vitamin C (ascorbic acid), a water-soluble essential vitamin, is well-known as an antioxidant and an essential substrate for several neutrophil functions. Because of (i) the importance of neutrophils in microbial control and (ii) the relatively low vitamin C level in neutrophils and in plasma during stress, vitamin C has been studied in sepsis (a life-threatening organ dysfunction from severe infection). Surprisingly, the supraphysiologic blood level of vitamin C (higher than 5 mM) after the high-dose intravenous vitamin C (HDIVC) for 4 days possibly induces the pro-oxidant effect in the extracellular space. As such, HDIVC demonstrates beneficial effects in sepsis which might be due to the impacts on an enhanced microbicidal activity through the improved activity indirectly via enhanced neutrophil functions and directly from the extracellular pro-oxidant effect on the organismal membrane. The concentration-related vitamin C properties are also observed in the neutrophil extracellular traps (NETs) formation as ascorbate inhibits NETs at 1 mM (or less) but facilitates NETs at 5 mM (or higher) concentration. The longer duration of HDIVC administration might be harmful in sepsis because NETs and pro-oxidants are partly responsible for sepsis-induced injuries, despite the possible microbicidal benefit. Despite the negative results in several randomized control trials, the short course HDIVC might be interesting to use in some selected groups, such as against anti-biotic resistant organisms. More studies on the proper use of vitamin C, a low-cost and widely available drug, in sepsis are warranted.
Collapse
|
4
|
Lisboa RV, de Oliveira FR, Quaresma TO, de Almeida RM, Ribeiro Oliveira RD, Junior PL. The Behaviour of Serum Survivin in Patients With Lupus Nephritis. Biomark Insights 2022; 17:11772719221131470. [PMID: 36311208 PMCID: PMC9597205 DOI: 10.1177/11772719221131470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 09/21/2022] [Indexed: 11/07/2022] Open
Abstract
Background: Systemic lupus erythematosus (SLE) is a chronic, multi phenotypic, autoimmune
inflammatory disease and renal involvement significantly worsens its
prognosis. Apoptosis dysregulation plays a key pathogenic role. Survivin, a
protein from the apoptosis inhibitors family, has been considered a
promising strategy in cancer therapy and evaluated as one of the regulatory
pathways in the scenario of immune-mediated disorders. Objective: This study aims to explore survivin behaviour in SLE patients with lupus
nephritis (LN), assessing its potential as a therapeutic and prognostic
biomarker. Methods: 297 SLE patients were classified based on the American College of
Rheumatology (ACR) 1997 criteria, from 2000 to 2015. In a cross-sectional
study, the serum level of survivin was measured by an ELISA test and
compared between 200 SLE individuals and healthy controls. In a longitudinal
cohort, 97 patients with active LN had the concentration of survinin
measured, before and after treatment with cyclophosphamide pulse
therapy. Results: The serum concentration of survivin was significantly lower in the SLE group
than in healthy controls, regardless of concomitant NL or disease activity.
The longitudinal evaluation revealed a significant reduction in survivin
serum level after treatment. However, survivin rates were not able to
discriminate groups that achieved remission from those that maintained
nephritis activity. Conclusion: Our study suggests that survivin levels in SLE patients are lower than in the
general population. Even so, its use as a biomarker in SLE seems limited,
not reflecting disease activity or response to LN treatment, as in other
contexts.
Collapse
Affiliation(s)
- Renata Valente Lisboa
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil,Renata Valente Lisboa, Division of
Rheumatology, Ribeirão Preto Medical School, University of São Paulo, Av.
Bandeirantes, 3900, Ribeirão Preto, São Paulo 14049-900, Brazil.
| | - Fabiola Reis de Oliveira
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil
| | - Thaise Oliveira Quaresma
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil
| | | | - Rene Donizeti Ribeiro Oliveira
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil
| | - Paulo Louzada Junior
- Ribeirão Preto Medical School, Ribeirão
Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil,Center of Research in Inflammatory
Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão
Preto, São Paulo, Brazil
| |
Collapse
|
5
|
Neutrophil extracellular traps accelerate vascular smooth muscle cell proliferation via Akt/CDKN1b/TK1 accompanying with the occurrence of hypertension. J Hypertens 2022; 40:2045-2057. [PMID: 35950975 PMCID: PMC9451946 DOI: 10.1097/hjh.0000000000003231] [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] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) can trigger pathological changes in vascular cells or vessel wall components, which are vascular pathological changes of hypertension. Therefore, we hypothesized that NETs would be associated with the occurrence of hypertension. METHODS To evaluate the relationship between NETs and hypertension, we evaluated both the NETs formation in spontaneously hypertensive rats (SHRs) and the blood pressure of mice injected phorbol-12-myristate-13-acetate (PMA) via the tail vein to induce NETs formation in arterial wall. Meanwhile, proliferation and cell cycle of vascular smooth muscle cells (VSMCs), which were co-cultured with NETs were assessed. In addition, the role of exosomes from VSMCs co-cultured with NETs on proliferation signaling delivery was assessed. RESULTS Formation of NETs increased in the arteries of SHR. PMA resulted in up-regulation expression of citrullinated Histone H3 (cit Histone H3, a NETs marker) in the arteries of mice accompanied with increasing of blood pressure. NET treatment significantly increased VSMCs count and accelerated G1/S transition in vitro . Cyclin-dependent kinase inhibitor 1b (CDKN1b) was down-regulated and Thymidine kinase 1 (TK1) was up-regulated in VSMCs. Exosomes from VSMCs co-cultured with NETs significantly accelerated the proliferation of VSMCs. TK1 was up-regulated in the exosomes from VSMCs co-cultured with NETs and in both the arterial wall and serum of mice with PMA. CONCLUSION NETs promote VSMCs proliferation via Akt/CDKN1b/TK1 and is related to hypertension development. Exosomes from VSMCs co-cultured with NETs participate in transferring the proliferation signal. These results support the role of NETs in the development of hypertension.
Collapse
|
6
|
Han T, Tang H, Lin C, Shen Y, Yan D, Tang X, Guo D. Extracellular traps and the role in thrombosis. Front Cardiovasc Med 2022; 9:951670. [PMID: 36093130 PMCID: PMC9452724 DOI: 10.3389/fcvm.2022.951670] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Thrombotic complications pose serious health risks worldwide. A significant change in our understanding of the pathophysiology of thrombosis has occurred since the discovery of extracellular traps (ETs) and their prothrombotic properties. As a result of immune cells decondensing chromatin into extracellular fibers, ETs promote thrombus formation by acting as a scaffold that activates platelets and coagulates them. The involvement of ETs in thrombosis has been reported in various thrombotic conditions including deep vein thrombosis (DVT), pulmonary emboli, acute myocardial infarction, aucte ischemic stroke, and abdominal aortic aneurysms. This review summarizes the existing evidence of ETs in human and animal model thrombi. The authors described studies showing the existence of ETs in venous or arterial thrombi. In addition, we studied potential novel therapeutic opportunities related to the resolution or prevention of thrombosis by targeting ETs.
Collapse
|
7
|
Platelet-Neutrophil Interactions and Thrombo-inflammatory Complications in Type 2 Diabetes Mellitus. CURRENT PATHOBIOLOGY REPORTS 2022. [DOI: 10.1007/s40139-022-00229-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Chen Z, Wei X, Dong S, Han F, He R, Zhou W. Challenges and Opportunities Associated With Platelets in Pancreatic Cancer. Front Oncol 2022; 12:850485. [PMID: 35494001 PMCID: PMC9039220 DOI: 10.3389/fonc.2022.850485] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/15/2022] [Indexed: 01/02/2023] Open
Abstract
Pancreatic cancer is one of the most common malignant tumors in the digestive system with a poor prognosis. Accordingly, better understanding of the molecular mechanisms and innovative therapies are warranted to improve the prognosis of this patient population. In addition to playing a crucial role in coagulation, platelets reportedly contribute to the growth, invasion and metastasis of various tumors, including pancreatic cancer. This narrative review brings together currently available evidence on the impact of platelets on pancreatic cancer, including the platelet-related molecular mechanisms of cancer promotion, pancreatic cancer fibrosis, immune evasion, drug resistance mechanisms, thrombosis, targeted platelet therapy, combined radiotherapy and chemotherapy treatment, platelet combined with nanotechnology treatment and potential applications of pancreatic cancer organoids. A refined understanding of the role of platelets in pancreatic cancer provides the foothold for identifying new therapeutic targets.
Collapse
Affiliation(s)
- Zhou Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaodong Wei
- Emergency Department, Gansu Provincial Hospital, Lanzhou, China
| | - Shi Dong
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Fangfang Han
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ru He
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
9
|
Platelets in COVID-19 disease: friend, foe, or both? Pharmacol Rep 2022; 74:1182-1197. [PMID: 36463349 PMCID: PMC9726679 DOI: 10.1007/s43440-022-00438-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 12/07/2022]
Abstract
Immuno-thrombosis of COVID-19 results in the activation of platelets and coagulopathy. Antiplatelet therapy has been widely used in COVID-19 patients to prevent thrombotic events. However, recent analysis of clinical trials does not support the major effects of antiplatelet therapy on mortality in hospitalized COVID-19 patients, despite the indisputable evidence for an increased risk of thrombotic complications in COVID-19 disease. This apparent paradox calls for an explanation. Platelets have an important role in sensing and orchestrating host response to infection, and several platelet functions related to host defense response not directly related to their well-known hemostatic function are emerging. In this paper, we aim to review the evidence supporting the notion that platelets have protective properties in maintaining endothelial barrier integrity in the course of an inflammatory response, and this role seems to be of particular importance in the lung. It might, thus, well be that the inhibition of platelet function, if affecting the protective aspect of platelet activity, might diminish clinical benefits resulting from the inhibition of the pro-thrombotic phenotype of platelets in immuno-thrombosis of COVID-19. A better understanding of the platelet-dependent mechanisms involved in the preservation of the endothelial barrier is necessary to design the antiplatelet therapeutic strategies that inhibit the pro-thrombotic activity of platelets without effects on the vaso-protective function of platelets safeguarding the pulmonary endothelial barrier during multicellular host defense in pulmonary circulation.
Collapse
|
10
|
Tzankov A, Bhattacharyya S, Kotlo K, Tobacman JK. Increase in Chondroitin Sulfate and Decline in Arylsulfatase B May Contribute to Pathophysiology of COVID-19 Respiratory Failure. Pathobiology 2021; 89:81-91. [PMID: 34788765 DOI: 10.1159/000519542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/06/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The potential role of accumulation of chondroitin sulfates (CSs) in the pathobiology of COVID-19 has not been examined. Accumulation may occur by increased synthesis or by decline in activity of the enzyme arylsulfatase B (ARSB; N-acetylgalactosamine-4-sulfatase) which requires oxygen for activity. METHODS Immunostaining of lung tissue from 28 patients who died due to COVID-19 infection was performed for CS, ARSB, and carbohydrate sulfotransferase (CHST)15. Measurements of mRNA expression of CHST15 and CHST11, sulfotransferase activity, and total sulfated glycosaminoglycans (GAGs) were determined in human vascular smooth muscle cells following angiotensin (Ang) II treatment. RESULTS CS immunostaining showed increase in intensity and distribution, and immunostaining of ARSB was diminished in COVID-19 compared to normal lung tissue. CHST15 immunostaining was prominent in vascular smooth muscle cells associated with diffuse alveolar damage due to COVID-19 or other causes. Expression of CHST15 and CHST11 which are required for synthesis of CSE and chondroitin 4-sulfate, total sulfated GAGs, and sulfotransferase activity was significantly increased following AngII exposure in vascular smooth muscle cells. Expression of Interleukin-6 (IL-6), a mediator of cytokine storm in COVID-19, was inversely associated with ARSB expression. DISCUSSION/CONCLUSION Decline in ARSB and resulting increases in CS may contribute to the pathobiology of COVID-19, as IL-6 does. Increased expression of CHSTs following activation of Ang-converting enzyme 2 may lead to buildup of CSs.
Collapse
Affiliation(s)
- Alexandar Tzankov
- Pathology, University Hospital Basel, Institute of Medical Genetics and Pathology, University of Basel, Basel, Switzerland
| | - Sumit Bhattacharyya
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Kumar Kotlo
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Joanne K Tobacman
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Jesse Brown VA Medical Center, Chicago, Illinois, USA
| |
Collapse
|
11
|
Jo HM, Ahn C, Kim H, Kang BT, Jeung EB, Yang MP. Effect of quercetin on formation of porcine neutrophil extracellular trap. Vet Immunol Immunopathol 2021; 241:110335. [PMID: 34627080 DOI: 10.1016/j.vetimm.2021.110335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 08/02/2021] [Accepted: 10/02/2021] [Indexed: 10/20/2022]
Abstract
Neutrophil extracellular trap (NET) formation is an immune response to the invasion of external microorganisms. Quercetin, a member of the flavonoid family found in fruits and vegetables, has been examined in multiple biological contexts. The objective of this study was to examine the effect of quercetin on porcine NET formation. We measured NET formation by peripheral blood polymorphonuclear cells (PMNs) using propidium iodide (PI) dye. The amount of tumor necrosis factor (TNF)-α in culture supernatants was quantified by ELISA, and TNF-α mRNA expression was measured by RT-PCR. Direct treatment of PMNs with quercetin did not affect NET formation; however, NET formation was inhibited by exposure to culture supernatant from peripheral blood mononuclear cells (PBMCs) treated with quercetin. By contrast, culture supernatant from PBMCs treated with lipopolysaccharide (LPS) induced high levels of NET formation of PMNs, and this effect was reduced by co-treatment with LPS and quercetin. In addition, treatment of PMNs with recombinant porcine (rp) TNF-α induced high levels of NET formation. PBMCs treated with LPS increased higher levels of TNF-α mRNA and protein, but this effect was weakened when they were co-treated with quercetin. These findings indicated that quercetin inhibits NET formation of PMNs by suppressing production of TNF-α from LPS-stimulated PBMCs. These results suggest that quercetin exerts an anti-inflammatory effect, mediated by down-regulation of TNF-α production from LPS-stimulated PBMCs, which inhibits NET formation in PMNs.
Collapse
Affiliation(s)
- Hyun-Min Jo
- Department of Veterinary Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Changhwan Ahn
- Department of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju Special Self Governing Province, 63243, Republic of Korea
| | - Hakhyun Kim
- Department of Veterinary Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Byeong-Teck Kang
- Department of Veterinary Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Eui-Bae Jeung
- Department of Veterinary Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Mhan-Pyo Yang
- Department of Veterinary Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
12
|
Zapponi KCS, Orsi FA, Cunha JLR, de Brito IR, Romano AVC, Bittar LF, De Paula EV, Penteado CF, Montalvão S, Annichino-Bizzacchi JM. Neutrophil activation and circulating neutrophil extracellular traps are increased in venous thromboembolism patients for at least one year after the clinical event. J Thromb Thrombolysis 2021; 53:30-42. [PMID: 34449018 PMCID: PMC8791881 DOI: 10.1007/s11239-021-02526-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2021] [Indexed: 12/23/2022]
Abstract
Neutrophil activation and neutrophil extracellular traps (NETs) have been associated with the pathogenesis of venous thromboembolism (VTE). Considering VTE-associated chronic sequelae, which suggest that some pathological mechanisms remain after the acute episode, we investigated whether neutrophil activation is increased in patients with a prior VTE at least one year before this investigation. Thirty-seven patients with prior VTE and 37 individuals with no history of VTE were included. Neutrophil activity was evaluated by the expression of the adhesive molecule activation-specific epitopes LFA-1 (CD11a) and MAC-1 (CD11b), chemotaxis, reactive oxygen species (ROS) and by MPO-DNA complexes as markers of NETs. The adhesive molecules sICAM-1 and sVCAM-1, involved in the cross talk between neutrophil and endothelial cells, were also evaluated. Patient neutrophils presented increased CD11a expression before and after TNF-α stimulus, whereas increased CD11b expression was observed only after TNF-α stimulus, as compared to controls. Neutrophil chemotaxis on both, basal state and after IL-8 stimulus, on circulating levels of sICAM-1 and sVCAM-1, and on MPO-DNA complexes were also increased in VTE patients. ROS release was similar between patients and controls. This is, to our knowledge, the first study to investigate neutrophil inflammatory activity in VTE patients a long period after an acute event (approximately 2 years). The results showed altered neutrophil activation patterns in these patients. While activated neutrophils can cause endothelial activation and injury, the activated endothelium can induce the release of NETs with consequent endothelial cytotoxicity, creating a vicious cycle of activation between neutrophils and endothelium that can lead to thrombosis.
Collapse
Affiliation(s)
- Kiara C. S. Zapponi
- Hematology and Hemotherapy Center, University of Campinas, Carlos Chagas street, 480, Campinas, 13083878 Brazil
| | - Fernanda A. Orsi
- Department of Clinical Pathology, School of Medical Sciences, University of Campinas, Campinas, SP Brazil
- Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - José Luiz R. Cunha
- Department of Clinical Pathology, School of Medical Sciences, University of Campinas, Campinas, SP Brazil
| | - Ingrid R. de Brito
- Department of Clinical Pathology, School of Medical Sciences, University of Campinas, Campinas, SP Brazil
| | - Anna Virginia C. Romano
- Hematology and Hemotherapy Center, University of Campinas, Carlos Chagas street, 480, Campinas, 13083878 Brazil
| | - Luis Fernando Bittar
- Hematology and Hemotherapy Center, University of Campinas, Carlos Chagas street, 480, Campinas, 13083878 Brazil
- Department of Clinical Pathology, School of Medical Sciences, University of Campinas, Campinas, SP Brazil
| | - Erich Vinicius De Paula
- Hematology and Hemotherapy Center, University of Campinas, Carlos Chagas street, 480, Campinas, 13083878 Brazil
| | - Carla F. Penteado
- Hematology and Hemotherapy Center, University of Campinas, Carlos Chagas street, 480, Campinas, 13083878 Brazil
| | - Silmara Montalvão
- Hematology and Hemotherapy Center, University of Campinas, Carlos Chagas street, 480, Campinas, 13083878 Brazil
| | | |
Collapse
|
13
|
Tabrizi ZA, Khosrojerdi A, Aslani S, Hemmatzadeh M, Babaie F, Bairami A, Shomali N, Hosseinzadeh R, Safari R, Mohammadi H. Multi-facets of neutrophil extracellular trap in infectious diseases: Moving beyond immunity. Microb Pathog 2021; 158:105066. [PMID: 34174356 DOI: 10.1016/j.micpath.2021.105066] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
Neutrophil extracellular traps (NETs) are networks of extracellular chromosomal DNA fibers, histones, and cytoplasmic granule proteins. The release of NET components from neutrophils is involved in the suppression of pathogen diffusion. Development of NETs around target microbes leads to disruption of the cell membrane, eventuating in kind of cell death that is called as NETosis. The very first step in the process of NETosis is activation of Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase upon signaling by innate immune receptors. Afterwards, produced Reactive oxygen species (ROS) trigger protein-arginine deiminase type 4, neutrophil elastase, and myeloperoxidase to generate decondensed chromatin and disrupted integrity of nuclear membrane. Subsequently, decondensed chromatin is mixed with several enzymes in the cytoplasm released from granules, leading to release of DNA and histones, and finally formation of NET. Several reports have indicated that NETosis might contribute to the immune responses through limiting the dissemination of microbial organisms. In this review, we discuss recent advances on the role of neutrophils, NETs, and their implications in the pathogenesis of microbial infections. Additionally, the prospective of the NET modulation as a therapeutic strategy to treat infectious diseases are clarified.
Collapse
Affiliation(s)
- Zahra Azimzadeh Tabrizi
- Department of Laboratory Sciences, School of Allied Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Arezou Khosrojerdi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Babaie
- Department of Immunology and Genetic, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran; Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Amir Bairami
- Department of Medical Parasitology and Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Roghaiyeh Safari
- Molecular and Cellular Epigenetics, GIGA, University of Liege, Sart-Tilman Liège, Belgium; Molecular and Cellular Biology, TERRA, Gembloux Agro-Bio Tech, University of Liege, Gembloux, Belgium
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
14
|
Blanch-Ruiz MA, Ortega-Luna R, Martínez-Cuesta MÁ, Álvarez Á. The Neutrophil Secretome as a Crucial Link between Inflammation and Thrombosis. Int J Mol Sci 2021; 22:4170. [PMID: 33920656 PMCID: PMC8073391 DOI: 10.3390/ijms22084170] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases are a leading cause of death. Blood-cell interactions and endothelial dysfunction are fundamental in thrombus formation, and so further knowledge of the pathways involved in such cellular crosstalk could lead to new therapeutical approaches. Neutrophils are secretory cells that release well-known soluble inflammatory signaling mediators and other complex cellular structures whose role is not fully understood. Studies have reported that neutrophil extracellular vesicles (EVs) and neutrophil extracellular traps (NETs) contribute to thrombosis. The objective of this review is to study the role of EVs and NETs as key factors in the transition from inflammation to thrombosis. The neutrophil secretome can promote thrombosis due to the presence of different factors in the EVs bilayer that can trigger blood clotting, and to the release of soluble mediators that induce platelet activation or aggregation. On the other hand, one of the main pathways by which NETs induce thrombosis is through the creation of a scaffold to which platelets and other blood cells adhere. In this context, platelet activation has been associated with the induction of NETs release. Hence, the structure and composition of EVs and NETs, as well as the feedback mechanism between the two processes that causes pathological thrombus formation, require exhaustive analysis to clarify their role in thrombosis.
Collapse
Affiliation(s)
- María Amparo Blanch-Ruiz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - María Ángeles Martínez-Cuesta
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| |
Collapse
|
15
|
Herster F, Karbach S, Chatterjee M, Weber ANR. Platelets: Underestimated Regulators of Autoinflammation in Psoriasis. J Invest Dermatol 2021; 141:1395-1403. [PMID: 33810836 DOI: 10.1016/j.jid.2020.12.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 01/01/2023]
Abstract
Platelets have long been known as mediators of hemostasis and, more recently, as mediators of thromboinflammation, although their physiopathological role has mostly been investigated in the context of disease of internal organs, such as liver and kidney, or systemic disorders. Of late, exciting recent data suggest that platelets may also play a role in inflammation at distal sites such as the skin: recent studies show that platelets, by engaging polymorphonuclear neutrophils (PMNs), contribute to local inflammation in the frequent skin disorder, psoriasis. In an experimental model, systemic depletion of platelets drastically attenuated skin inflammation by preventing PMN infiltration of the skin. A broader role of platelets in different types of skin inflammation is therefore likely, and in this paper, we specifically review recent advances in psoriasis. Special emphasis is given to the crosstalk with systemic platelet effects, which may be of interest in psoriasis-related cardiovascular comorbidities. Furthermore, we discuss the potential for platelet-centered interventions in the therapy for psoriasis.
Collapse
Affiliation(s)
- Franziska Herster
- Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany; Department of Molecular Oncology, Robert Bosch Centrum für Tumorerkrankungen (RBCT), Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Susanne Karbach
- Center for Cardiology - Cardiology I, University Medical Center Mainz and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
16
|
The molecular basis of immune-based platelet disorders. Clin Sci (Lond) 2021; 134:2807-2822. [PMID: 33140828 DOI: 10.1042/cs20191101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/12/2020] [Accepted: 10/23/2020] [Indexed: 12/17/2022]
Abstract
Platelets have a predominant role in haemostasis, the maintenance of blood volume and emerging roles as innate immune cells, in wound healing and in inflammatory responses. Platelets express receptors that are important for platelet adhesion, aggregation, participation in inflammatory responses, and for triggering degranulation and enhancing thrombin generation. They carry a cargo of granules bearing enzymes, adhesion molecules, growth factors and cytokines, and have the ability to generate reactive oxygen species. The platelet is at the frontline of a host of cellular responses to invading pathogens, injury, and infection. Perhaps because of this intrinsic responsibility of a platelet to rapidly respond to thrombotic, pathological and immunological factors as part of their infantry role; platelets are susceptible to targeted attack by the adaptive immune system. Such attacks are often transitory but result in aberrant platelet activation as well as significant loss of platelet numbers and platelet function, paradoxically leading to elevated risks of both thrombosis and bleeding. Here, we discuss the main molecular events underlying immune-based platelet disorders with specific focus on events occurring at the platelet surface leading to activation and clearance.
Collapse
|
17
|
Shahar M, Szalat A, Rosen H. Pathogenic Stress Induces Human Monocyte to Express an Extracellular Web of Tunneling Nanotubes. Front Immunol 2021; 12:620734. [PMID: 33679763 PMCID: PMC7933571 DOI: 10.3389/fimmu.2021.620734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Actin-based tunneling nanotubes are a means of intercellular communication between remote cells. In the last decade, this type of nanotube was described in a wide variety of cell types and it became widely accepted that communication through these nanotubes is related to response to environmental changes. Few reports, however, are available regarding the expression of similar nanotubes in vivo or in primary cells. Moreover, the functional significance of this intercellular communication for health and disease is largely unknown. In this context, and as a first step in unraveling these questions, we examined the formation of similar nanotubes in primary peripheral human monocytes. To that end, we combined the use of a live cell imaging system along with advanced methods of fluorescent and scanning electron microscopy. This experimental approach reveals for the first time that the bacterial lipopolysaccharide endotoxin induces a transient expression of an unexpected abundance of actin-based tunneling nanotubes associated with vesicles. In addition, it was found that a similar response can be achieved by treating human monocytes with various bacterial and yeast membrane components, as well as with a viral component analog. In all these cases, this response is mediated by distinct complexes of toll-like receptors. Therefore, we suggest that the observed phenomena are related to a broad type of monocyte pathogen response, and raise the possibility that the phenomena described above may be involved in many clinical situations related to inflammation as a new topic of study.
Collapse
Affiliation(s)
- Michal Shahar
- The Department of Microbiology and Molecular Genetics, Institute for Medical Research-Israel-Canada, Hebrew University - Hadassah Medical Center, Jerusalem, Israel
| | - Auryan Szalat
- Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Haim Rosen
- The Kuvin Center for the Study of Infectious and Tropical Diseases, Institute for Medical Research-Israel-Canada, Hebrew University - Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
18
|
Platelet CXCL4 mediates neutrophil extracellular traps formation in ANCA-associated vasculitis. Sci Rep 2021; 11:222. [PMID: 33420306 PMCID: PMC7794527 DOI: 10.1038/s41598-020-80685-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Neutrophils form neutrophil extracellular traps (NETs), which are involved in the pathogenesis of ANCA-associated vasculitis (AAV). Recent reports suggest that platelets stimulated via toll-like receptor (TLR) pathways can induce NETs formation. However, the mechanism underlying the involvement of platelets in NETs formation in AAV is unknown. We investigated the role of platelets in the pathogenesis of AAV. Platelets from AAV patients and healthy controls (HCs) were co-cultured with peripheral neutrophils, and NETs formation was visualized and quantified. The expression levels of TLRs on platelets were examined by flow cytometry. Platelets were treated with a TLR agonist, platelet-derived humoral factor, CXCL4 (platelet factor 4: PF4), and/or anti-CXCL4 antibody to investigate the effects of TLR–CXCL4 signaling on NETs formation. Platelets from AAV significantly upregulated NETs formation in vitro. Flow cytometric analysis revealed that the proportion of TLR9 positive platelets was significantly higher in AAV than HCs. CXCL4 released from TLR9 agonist-stimulated platelets was significantly enhanced in AAV, which subsequently increased NETs formation. Further, neutralizing anti-CXCL4 antibody significantly inhibited NETs formation enhanced by platelets from AAV. TLR9 signaling and CXCL4 release underlie the key role that platelets play in NETs formation in the pathogenesis of AAV.
Collapse
|
19
|
Miró J, Marín H, Catalán J, Papas M, Gacem S, Yeste M. Seminal Plasma, Sperm Concentration, and Sperm-PMN Interaction in the Donkey: An In Vitro Model to Study Endometrial Inflammation at Post-Insemination. Int J Mol Sci 2020; 21:ijms21103478. [PMID: 32423134 PMCID: PMC7278951 DOI: 10.3390/ijms21103478] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 12/16/2022] Open
Abstract
In the donkey, artificial insemination (AI) with frozen-thawed semen is associated with low fertility rates, which could be partially augmented through adding seminal plasma (SP) and increasing sperm concentration. On the other hand, post-AI endometrial inflammation in the jenny is significantly higher than in the mare. While previous studies analyzed this response through recovering Polymorphonuclear Neutrophils (PMN) from uterine washings, successive lavages can detrimentally impact the endometrium, leading to fertility issues. For this reason, the first set of experiments in this work intended to set an in vitro model through harvesting PMN from the peripheral blood of jennies. Thereafter, how PMN, which require a triggering agent like formyl-methionyl-leucyl-phenylalanine (FMLP) to be activated, are affected by donkey semen was interrogated. Finally, we tested how four concentrations of spermatozoa (100 × 106, 200 × 106, 500 × 106 and 1000 × 106 spermatozoa/mL) affected their interaction with PMN. We observed that semen, which consists of sperm and SP, is able to activate PMN. Whereas there was a reduced percentage of spermatozoa phagocytosed by PMN, most remained attached on the PMN surface or into a surrounding halo. Spermatozoa not attached to PMN were viable, and most of those bound to PMN were also viable and showed high tail beating. Finally, only sperm concentrations higher than 500 × 106 spermatozoa/mL showed free sperm cells after 3 h of incubation, and percentages of spermatozoa not attached to PMN were higher at 3 h than at 1 h, exhibiting high motility. We can thus conclude that semen activates PMN in the donkey, and that the percentage of spermatozoa phagocytosed by PMN is low. Furthermore, because percentages of spermatozoa not attached to PMN were higher after 3 h than after 1 h of incubation, we suggest that PMN-sperm interaction plays an instrumental role in the reproductive strategy of the donkey.
Collapse
Affiliation(s)
- Jordi Miró
- Equine Reproduction Service, Department of Animal Medicine and Surgery, Faculty of Veterinary Sciences, Autonomous University of Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (H.M.); (J.C.); (M.P.); (S.G.)
- Correspondence: ; Tel.: +34-93-5814273
| | - Henar Marín
- Equine Reproduction Service, Department of Animal Medicine and Surgery, Faculty of Veterinary Sciences, Autonomous University of Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (H.M.); (J.C.); (M.P.); (S.G.)
| | - Jaime Catalán
- Equine Reproduction Service, Department of Animal Medicine and Surgery, Faculty of Veterinary Sciences, Autonomous University of Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (H.M.); (J.C.); (M.P.); (S.G.)
| | - Marion Papas
- Equine Reproduction Service, Department of Animal Medicine and Surgery, Faculty of Veterinary Sciences, Autonomous University of Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (H.M.); (J.C.); (M.P.); (S.G.)
| | - Sabrina Gacem
- Equine Reproduction Service, Department of Animal Medicine and Surgery, Faculty of Veterinary Sciences, Autonomous University of Barcelona, E-08193 Bellaterra (Cerdanyola del Vallès), Spain; (H.M.); (J.C.); (M.P.); (S.G.)
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, E-17003 Girona, Spain;
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, E-17003 Girona, Spain
| |
Collapse
|
20
|
P2Y 12 Inhibition beyond Thrombosis: Effects on Inflammation. Int J Mol Sci 2020; 21:ijms21041391. [PMID: 32092903 PMCID: PMC7073040 DOI: 10.3390/ijms21041391] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/18/2022] Open
Abstract
The P2Y12 receptor is a key player in platelet activation and a major target for antithrombotic drugs. The beneficial effects of P2Y12 receptor antagonists might, however, not be restricted to the primary and secondary prevention of arterial thrombosis. Indeed, it has been established that platelet activation also has an essential role in inflammation. Additionally, nonplatelet P2Y12 receptors present in immune cells and vascular smooth muscle cells might be effective players in the inflammatory response. This review will investigate the biological and clinical impact of P2Y12 receptor inhibition beyond its platelet-driven antithrombotic effects, focusing on its anti-inflammatory role. We will discuss the potential molecular and cellular mechanisms of P2Y12-mediated inflammation, including cytokine release, platelet–leukocyte interactions and neutrophil extracellular trap formation. Then we will summarize the current evidence on the beneficial effects of P2Y12 antagonists during various clinical inflammatory diseases, especially during sepsis, acute lung injury, asthma, atherosclerosis, and cancer.
Collapse
|
21
|
Wirestam L, Arve S, Linge P, Bengtsson AA. Neutrophils-Important Communicators in Systemic Lupus Erythematosus and Antiphospholipid Syndrome. Front Immunol 2019; 10:2734. [PMID: 31824510 PMCID: PMC6882868 DOI: 10.3389/fimmu.2019.02734] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS) are two autoimmune diseases that can occur together or separately. Insights into the pathogenesis have revealed similarities, such as development of autoantibodies targeting subcellular antigens as well as a shared increased risk of cardiovascular morbidity, potentially due to mutual pathologic mechanisms. In this review, we will address the evidence implicating neutrophils in the pathogenesis of these conditions, highlighting their shared features. The neutrophil is the most abundant leukocyte, recognized for its role in infectious and inflammatory diseases, but dysregulation of neutrophil effector functions, including phagocytosis, oxidative burst and formation of neutrophil extracellular traps (NETs) may also contribute to an autoimmune process. The phenotype of neutrophils in SLE and APS differs from neutrophils of healthy individuals, where neutrophils in SLE and APS are activated and prone to aggregate. A specific subset of low-density neutrophils with different function compared to normal-density neutrophils can also be found within the peripheral blood mononuclear cell (PBMC) fraction after density gradient centrifugation of whole blood. Neutrophil phagocytosis is required for regular clearance of cell remnants and nuclear material. Reactive oxygen species (ROS) released by neutrophils during oxidative burst are important for immune suppression and impairment of ROS production is seen in SLE. NETs mediate pathology in both SLE and APS via several mechanisms, including exposure of autoantigens, priming of T-cells and activation of autoreactive B-cells. NETs are also involved in cardiovascular events by forming a pro-thrombotic scaffolding surface. Lastly, neutrophils communicate with other cells by producing cytokines, such as Interferon (IFN) -α, and via direct cell-cell contact. Physiological neutrophil effector functions are necessary to prevent autoimmunity, but in SLE and APS these are altered.
Collapse
Affiliation(s)
- Lina Wirestam
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sabine Arve
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Petrus Linge
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Anders A Bengtsson
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Serebrovska ZO, Serebrovska TV, Kholin VA, Tumanovska LV, Shysh AM, Pashevin DA, Goncharov SV, Stroy D, Grib ON, Shatylo VB, Bachinskaya NY, Egorov E, Xi L, Dosenko VE. Intermittent Hypoxia-Hyperoxia Training Improves Cognitive Function and Decreases Circulating Biomarkers of Alzheimer's Disease in Patients with Mild Cognitive Impairment: A Pilot Study. Int J Mol Sci 2019; 20:E5405. [PMID: 31671598 PMCID: PMC6862463 DOI: 10.3390/ijms20215405] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) affects not only the central nervous system, but also peripheral blood cells including neutrophils and platelets, which actively participate in pathogenesis of AD through a vicious cycle between platelets aggregation and production of excessive amyloid beta (Aβ). Platelets adhesion on amyloid plaques also increases the risk of cerebral microcirculation disorders. Moreover, activated platelets release soluble adhesion molecules that cause migration, adhesion/activation of neutrophils and formation of neutrophil extracellular traps (NETs), which may damage blood brain barrier and destroy brain parenchyma. The present study examined the effects of intermittent hypoxic-hyperoxic training (IHHT) on elderly patients with mild cognitive impairment (MCI), a precursor of AD. Twenty-one participants (age 51-74 years) were divided into three groups: Healthy Control (n = 7), MCI+Sham (n = 6), and MCI+IHHT (n = 8). IHHT was carried out five times per week for three weeks (total 15 sessions). Each IHHT session consisted of four cycles of 5-min hypoxia (12% FIO2) and 3-min hyperoxia (33% FIO2). Cognitive parameters, Aβ and amyloid precursor protein (APP) expression, microRNA 29, and long non-coding RNA in isolated platelets as well as NETs in peripheral blood were investigated. We found an initial decline in cognitive function indices in both MCI+Sham and MCI+IHHT groups and significant correlations between cognitive test scores and the levels of circulating biomarkers of AD. Whereas sham training led to no change in these parameters, IHHT resulted in the improvement in cognitive test scores, along with significant increase in APP ratio and decrease in Aβ expression and NETs formation one day after the end of three-week IHHT. Such effects on Aβ expression and NETs formation remained more pronounced one month after IHHT. In conclusion, our results from this pilot study suggested a potential utility of IHHT as a new non-pharmacological therapy to improve cognitive function in pre-AD patients and slow down the development of AD.
Collapse
Affiliation(s)
- Zoya O Serebrovska
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.
| | | | - Viktor A Kholin
- Department of Age Physiology and Pathology of Nervous System, Chebotarev Institute of Gerontology NAMS of Ukraine, Kyiv 04114, Ukraine.
| | - Lesya V Tumanovska
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.
| | - Angela M Shysh
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.
| | - Denis A Pashevin
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.
| | - Sergii V Goncharov
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.
| | - Dmytro Stroy
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.
| | - Oksana N Grib
- Department of Clinical Physiology and Pathology of Internal Organs, Chebotarev Institute of Gerontology NAMS of Ukraine, Kyiv 04114, Ukraine.
| | - Valeriy B Shatylo
- Department of Clinical Physiology and Pathology of Internal Organs, Chebotarev Institute of Gerontology NAMS of Ukraine, Kyiv 04114, Ukraine.
| | - Natalia Yu Bachinskaya
- Department of Age Physiology and Pathology of Nervous System, Chebotarev Institute of Gerontology NAMS of Ukraine, Kyiv 04114, Ukraine.
| | - Egor Egorov
- CellAir Constructions GmbH, Schorndorf 73614, Germany.
| | - Lei Xi
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA 23298-0204, USA.
| | - Victor E Dosenko
- Department of General and Molecular Pathophysiology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine.
| |
Collapse
|
23
|
Hook JS, Cao M, Potera RM, Alsmadi NZ, Schmidtke DW, Moreland JG. Nox2 Regulates Platelet Activation and NET Formation in the Lung. Front Immunol 2019; 10:1472. [PMID: 31338092 PMCID: PMC6626916 DOI: 10.3389/fimmu.2019.01472] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022] Open
Abstract
The mortality rate of patients with critical illness has decreased significantly over the past two decades, but the rate of decline has slowed recently, with organ dysfunction as a major driver of morbidity and mortality. Among patients with the systemic inflammatory response syndrome (SIRS), acute lung injury is a common component with serious morbidity. Previous studies in our laboratory using a murine model of SIRS demonstrated a key role for NADPH oxidase 2 (Nox2)-derived reactive oxygen species in the resolution of inflammation. Nox2-deficient (gp91phox−/y) mice develop profound lung injury secondary to SIRS and fail to resolve inflammation. Alveolar macrophages from gp91phox−/y mice express greater levels of chemotactic and pro-inflammatory factors at baseline providing evidence that Nox2 in alveolar macrophages is critical for homeostasis. Based on the lung pathology with increased thrombosis in gp91phox−/y mice, and the known role of platelets in the inflammatory process, we hypothesized that Nox2 represses platelet activation. In the mouse model, we found that platelet-derived chemokine (C-X-C motif) ligand 4 (CXCL4) and CXCL7 were increased in the bronchoalveolar fluid of gp91phox−/y mice at baseline and 24 h post intraperitoneal zymosan-induced SIRS consistent with platelet activation. Activated platelets interact with leukocytes via P-selectin glycoprotein ligand 1 (PSGL-1). Within 2 h of SIRS induction, alveolar neutrophil PSGL-1 expression was higher in gp91phox−/y mice. Platelet-neutrophil interactions were decreased in the peripheral blood of gp91phox−/y mice consistent with movement of activated platelets to the lung of mice lacking Nox2. Based on the severe lung pathology and the role of platelets in the formation of neutrophil extracellular traps (NETs), we evaluated NET production. In contrast to previous studies demonstrating Nox2-dependent NET formation, staining of lung sections from mice 24 h post zymosan injection revealed a large number of citrullinated histone 3 (H3CIT) and myeloperoxidase positive cells consistent with NET formation in gp91phox−/y mice that was virtually absent in WT mice. In addition, H3CIT protein expression and PAD4 activity were higher in the lung of gp91phox−/y mice post SIRS induction. These results suggest that Nox2 plays a critical role in maintaining homeostasis by regulating platelet activation and NET formation in the lung.
Collapse
Affiliation(s)
- Jessica S Hook
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States
| | - Mou Cao
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States
| | - Renee M Potera
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States
| | - Nesreen Z Alsmadi
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - David W Schmidtke
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Jessica G Moreland
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States.,Department of Microbiology, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
24
|
Zucoloto AZ, Jenne CN. Platelet-Neutrophil Interplay: Insights Into Neutrophil Extracellular Trap (NET)-Driven Coagulation in Infection. Front Cardiovasc Med 2019; 6:85. [PMID: 31281822 PMCID: PMC6595231 DOI: 10.3389/fcvm.2019.00085] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/06/2019] [Indexed: 01/06/2023] Open
Abstract
Well established for their central role in hemostasis, platelets have increasingly been appreciated as immune cells in recent years. This emerging role should not come as a surprise as the central immune cells of invertebrates, hemocytes, are able to phagocytose, secrete soluble mediators and promote coagulation of hemolymph, blurring the line between immunity and hemostasis. The undeniable evolutionary link between coagulation and immunity becomes even clearer as the role of platelets in inflammation is better understood. Platelets exert a range of immune-related functions, many of which involve an intimate interplay with leukocytes. Platelets promote leukocyte recruitment via endothelial activation and can serve as “landing pads” for leukocytes, facilitating cellular adhesion in vascular beds devoid of classic adhesion molecules. Moreover, platelets enhance leukocyte function both through direct interactions and through release of soluble mediators. Among neutrophil-platelets interactions, the modulation of neutrophil extracellular traps (NETs) is of great interest. Platelets have been shown to induce NET formation; and, in turn, NET components further regulate platelet and neutrophil function. While NETs have been shown to ensnare and kill pathogens, they also initiate coagulation via thrombin activation. In fact, increased NET formation has been associated with hypercoagulability in septic patients as well as in chronic vascular disorders. This review will delve into current knowledge of platelet-neutrophil interactions, with a focus on NET-driven coagulation, in the context of infectious diseases. A better understanding of these mechanisms will shed a light on the therapeutic potential of uncoupling immunity and coagulation through targeting of NETs.
Collapse
Affiliation(s)
- Amanda Z Zucoloto
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, The University of Calgary, Calgary, AB, Canada
| | - Craig N Jenne
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, The University of Calgary, Calgary, AB, Canada
| |
Collapse
|
25
|
Zhang Y, Wang C, Yu M, Zhao X, Du J, Li Y, Jing H, Dong Z, Kou J, Bi Y, Novakovic VA, Zhou J, Shi J. Neutrophil extracellular traps induced by activated platelets contribute to procoagulant activity in patients with colorectal cancer. Thromb Res 2019; 180:87-97. [PMID: 31271975 DOI: 10.1016/j.thromres.2019.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/03/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022]
Abstract
Patients with colorectal cancer (CRC) are at increased risk of venous thrombosis, but the precise mechanisms of thrombogenesis in CRC remain largely unknown. We aimed to identify the novel role of neutrophil extracellular traps (NETs) in the induction of procoagulant activity (PCA) in CRC, and to evaluate its interactions with platelets and endothelial cells (ECs). In this study, we first showed that the levels of NETs in the peripheral blood of CRC patients were increased in parallel with cancer progression and reached significance in stage II patients compared to healthy subjects. In addition, neutrophils from CRC patients were more prone to produce NETs, resulting in shortened coagulation time, significantly increased thrombin-antithrombin (TAT) complexes and fibrin fibrils compared to healthy controls. Furthermore, platelets from CRC patients stimulated healthy neutrophils to extrude NETs, which could be inhibited by the depletion of HMGB1. Conversely, NETs from CRC patients could also induce the exposure of PS on platelets, leading to markedly enhanced PCA. Importantly, ECs were also converted to a procoagulant phenotype when exposed to NETs from CRC patients. The PCA of NETs-activated platelets or ECs could be inhibited either by the cleavage of NETs with DNase1 or the blockage of histone with activated protein C (APC). Our results reveal the complex interactions between neutrophils, platelets and ECs and their potential role in the hypercoagulable state in CRC. We propose that NETs may provide new therapeutic targets to combat the thrombotic consequences of CRC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Chunxu Wang
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Muxin Yu
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Xinyi Zhao
- Department of Cardiology of the Second Hospital, Harbin Medical University, 148 Baojian Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Jingwen Du
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Yueyue Li
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Haijiao Jing
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Zengxiang Dong
- Department of Cardiology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Junjie Kou
- Department of Cardiology of the Second Hospital, Harbin Medical University, 148 Baojian Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Yayan Bi
- Department of Cardiology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China
| | - Valerie A Novakovic
- Departments of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, 1400 VFW Parkway, West Roxbury, Boston, MA, USA
| | - Jin Zhou
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China.
| | - Jialan Shi
- Department of Hematology of the First Hospital, Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province, China; Departments of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, 1400 VFW Parkway, West Roxbury, Boston, MA, USA.
| |
Collapse
|
26
|
Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res 2019; 29:347-364. [PMID: 30948788 PMCID: PMC6796845 DOI: 10.1038/s41422-019-0164-5] [Citation(s) in RCA: 1451] [Impact Index Per Article: 290.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
Cells may die from accidental cell death (ACD) or regulated cell death (RCD). ACD is a biologically uncontrolled process, whereas RCD involves tightly structured signaling cascades and molecularly defined effector mechanisms. A growing number of novel non-apoptotic forms of RCD have been identified and are increasingly being implicated in various human pathologies. Here, we critically review the current state of the art regarding non-apoptotic types of RCD, including necroptosis, pyroptosis, ferroptosis, entotic cell death, netotic cell death, parthanatos, lysosome-dependent cell death, autophagy-dependent cell death, alkaliptosis and oxeiptosis. The in-depth comprehension of each of these lethal subroutines and their intercellular consequences may uncover novel therapeutic targets for the avoidance of pathogenic cell loss.
Collapse
Affiliation(s)
- Daolin Tang
- The Third Affiliated Hospital, Protein Modification and Degradation Lab, School of Basic Medical Sciences, Guangzhou Medical University, 510510, Guangzhou, Guangdong, China.
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tom Vanden Berghe
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, 9052, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
- Laboratory of Pathophysiology, Faculty of Biomedical Sciences, University of Antwerp, 2610, Wilrijk, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, 9052, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
- Methusalem program, Ghent University, 9000, Ghent, Belgium
| | - Guido Kroemer
- Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France.
- Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, 75006, Paris, France.
- Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.
- Université Pierre et Marie Curie, 75006, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800, Villejuif, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015, Paris, France.
- Department of Women's and Children's Health, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
27
|
Kustanovich A, Schwartz R, Peretz T, Grinshpun A. Life and death of circulating cell-free DNA. Cancer Biol Ther 2019; 20:1057-1067. [PMID: 30990132 PMCID: PMC6606043 DOI: 10.1080/15384047.2019.1598759] [Citation(s) in RCA: 326] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/24/2019] [Accepted: 03/12/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor-specific, circulating cell-free DNA in liquid biopsies is a promising source of biomarkers for minimally invasive serial monitoring of treatment responses in cancer management. We will review the current understanding of the origin of circulating cell-free DNA and different forms of DNA release (including various types of cell death and active secretion processes) and clearance routes. The dynamics of extracellular DNA in blood during therapy and the role of circulating DNA in pathophysiological processes (tumor-associated inflammation, NETosis, and pre-metastatic niche development) provide insights into the mechanisms that contribute to tumor development and metastases formation. Better knowledge of circulating tumor-specific cell-free DNA could facilitate the development of new therapeutic and diagnostic options for cancer management.
Collapse
Affiliation(s)
- Anatoli Kustanovich
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ruth Schwartz
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tamar Peretz
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Albert Grinshpun
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
28
|
Influence of Interleukin-8 and Neutrophil Extracellular Trap (NET) Formation in the Tumor Microenvironment: Is There a Pathogenic Role? J Immunol Res 2019; 2019:6252138. [PMID: 31093511 PMCID: PMC6481028 DOI: 10.1155/2019/6252138] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/28/2019] [Indexed: 02/06/2023] Open
Abstract
In this review, we will highlight several studies that revolve around interleukin-8 (IL-8) and show the multiple facets that could take in the tumor microenvironment. Chemokines that attract neutrophils (to a large extent, IL-8) can have a bimodal behavior inducing the migration of them in the first place and later favoring the formation of NETs in the place of emission focus of the chemokine. Also, this mechanism occurs when neutrophils migrate to tumor cells and where the extrusion of NETs in the tumor is observed. A possible participation of NETs in cancer progression was considered; however, until now, it is difficult to decide if NETosis plays a pro- or antitumor role, although it is necessary to emphasize that there is more experimentation focused on the protumorigenic aspect of the NETs. The formation of NETs has a relevant role in the inhibition of the immune response against the tumor generated by neutrophils and in turn favoring the processes involved in the development of tumor metastasis. It is striking that we do not have more complete information about the effects of circulating chemokines on neutrophils in cancer patients and hence the suitability of this review. No one has observed to date the impact that it could have on other cell populations to inhibit the arrival of neutrophils and the formation/elimination of NETs. However, the extent to which NETs affect the function of other cells of the immune system in the tumor context has not been directly demonstrated. It is necessary to identify possible combinations of immunotherapy that involve the modulation of neutrophil activity with other strategies (immunomodulatory antibodies or adoptive cell therapy). Therefore, knowing the mechanisms by which tumors take advantage of this ability of neutrophils to form NETs is very important in the search for antitumor therapies and thus be able to take advantage of the possible immunotherapeutic combinations that we currently have in clinical practice.
Collapse
|
29
|
Elia E, Montecucco F, Portincasa P, Sahebkar A, Mollazadeh H, Carbone F. Update on pathological platelet activation in coronary thrombosis. J Cell Physiol 2018; 234:2121-2133. [PMID: 30317596 DOI: 10.1002/jcp.27575] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/17/2018] [Indexed: 12/19/2022]
Abstract
Although coronary thrombosis (CT) is integral to cardiovascular outcomes, the underlying pathophysiological mechanisms remain unclear. CT may occur in case of atherosclerotic plaque erosion/rupture, or even after stenting implantation. Platelets (PLT) activation is the keystone of atherothrombosis and depends on many dysregulated elements, including endothelial dysfunction, oxidized lipoproteins, and immune response. Besides the classical view of PLT as an effector of hemostatic response, a new repertoire of PLT activities is emerging. PLT lipidome oxidation is a self-maintaining process which promotes PLT reactivity, coagulation cascade, and inflammatory cell activation. PLT-innate immune cell interaction is also sustained by neutrophil extracellular traps and NLRP3 inflammasome pathways. Other noteworthy emerging mechanisms are implicated in the crosstalk between PLT and surrounding cells. Especially, microvesicles (MVs) released from PLT may extend their signaling network far beyond the classical cell-cell interactions. Moreover, the recognition of noncoding RNA in PLT MVs introduce another layer of complexity in terms of intercellular signaling by a direct regulation of messenger RNA profile and gene expression in the recipient cells. The aim of this narrative review is to update the recent advance in CT and intracoronary stent thrombosis, including causal factors and potential translation of experimental evidence into the clinical setting.
Collapse
Affiliation(s)
- Edoardo Elia
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy.,Department of Internal Medicine, First Clinic of Internal Medicine, Ospedale Policlinico San Martino, 10 Largo Benzi, Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, Clinica Medica "A. Murri," University of Bari Medical School, Bari, Italy
| | - Amirhossein Sahebkar
- Department of Pharmaceutical Biotechnology, Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Federico Carbone
- Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
30
|
Matsuhisa A, Okui A, Horiuchi Y. [Viewing sepsis and autoimmune disease in relation with infection and NETs-formation]. Nihon Saikingaku Zasshi 2018; 73:171-191. [PMID: 29863035 DOI: 10.3412/jsb.73.171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Neutrophil has been widely recognized as body's first line of defence against pathogens. NETosis was first reported in 2004 as a programmed cell death of neutrophil and distinguished from apoptosis and necrosis. This phenomenon has been already observed in both basic and clinical research. NETosis is induced by various stimulants such as PMA, IL-8, DAMPs/PAMPs, bacteria, and antigen-antibody complex including self-antibody such as ANCA. It is known that there are two types of NETosis following bacterial infections. Although both of them have the ability to capture and kill bacteria, they also damage the host tissues. The inhibition of the NETs-related enzymes prevents the NETs formation at that time. The production of O2- from respiratory burst of neutrophils triggers NETs formation. In the first type of NETosis, neutrophils are completely collapsed, while in the second type, they maintain the morphology and the ability of phagocytosis. However, bacteria can escape from NETs by degrading NETs with their secreting nucleases. Thus the animal models of infection, using these bacteria, oftentimes suffer from severe infectious diseases. Human CGD (Chronic Granulomatosis Disease) patients who do not have Nox2 are immunocompromised, and highly susceptible to infection due to the defect of NETs formation. On the other hand, SLE patients are unable to break down the NETs as their serum inhibits the DNase1 activity, which results in autoantibody generation against NETs as well as self-DNA. It is getting clear that there is a relationship between inflammatory diseases, including infectious diseases, Sepsis and autoimmune diseases, and NETs. Therefore, it is important to re-evaluate the inflammatory disorders from NETs' perspective, and to incorporate the emerging concepts for better understanding the mechanisms involved.
Collapse
Affiliation(s)
- Akio Matsuhisa
- Medical Device & Deagnostic Dept., Fuso Pharmaceutical Industries, Ltd
| | - Akira Okui
- Research & Development Center, Fuso Pharmaceutical Industries, Ltd
| | | |
Collapse
|
31
|
Wang Y, Ouyang Y, Liu B, Ma X, Ding R. Platelet activation and antiplatelet therapy in sepsis: A narrative review. Thromb Res 2018; 166:28-36. [DOI: 10.1016/j.thromres.2018.04.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 12/31/2022]
|
32
|
Caneparo V, Landolfo S, Gariglio M, De Andrea M. The Absent in Melanoma 2-Like Receptor IFN-Inducible Protein 16 as an Inflammasome Regulator in Systemic Lupus Erythematosus: The Dark Side of Sensing Microbes. Front Immunol 2018; 9:1180. [PMID: 29892303 PMCID: PMC5985366 DOI: 10.3389/fimmu.2018.01180] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/11/2018] [Indexed: 12/22/2022] Open
Abstract
Absent in melanoma 2 (AIM2)-like receptors (ALRs) are a newly characterized class of pathogen recognition receptors (PRRs) involved in cytosolic and nuclear pathogen DNA recognition. In recent years, two ALR family members, the interferon (IFN)-inducible protein 16 (IFI16) and AIM2, have been linked to the pathogenesis of various autoimmune diseases, among which systemic lupus erythematosus (SLE) has recently gained increasing attention. SLE patients are indeed often characterized by constitutively high serum IFN levels and increased expression of IFN-stimulated genes due to an abnormal response to pathogens and/or incorrect self-DNA recognition process. Consistently, we and others have shown that IFI16 is overexpressed in a wide range of autoimmune diseases where it triggers production of specific autoantibodies. In addition, evidence from mouse models supports a model whereby ALRs are required for IFN-mediated host response to both exogenous and endogenous DNA. Following interaction with cytoplasmic or nuclear nucleic acids, ALRs can form a functional inflammasome through association with the adaptor ASC [apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD)] and with procaspase-1. Importantly, inflammasome-mediated upregulation of IL-1β and IL-18 production positively correlates with SLE disease severity. Therefore, targeting ALR sensors and their downstream pathways represents a promising alternative therapeutic approach for SLE and other systemic autoimmune diseases.
Collapse
Affiliation(s)
- Valeria Caneparo
- Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin, Italy.,Virology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Department of Translational Medicine, Novara Medical School, Novara, Italy.,Intrinsic Immunity Unit, CAAD - Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| | - Santo Landolfo
- Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin, Italy
| | - Marisa Gariglio
- Virology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Department of Translational Medicine, Novara Medical School, Novara, Italy.,Intrinsic Immunity Unit, CAAD - Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| | - Marco De Andrea
- Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin, Italy.,Virology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Department of Translational Medicine, Novara Medical School, Novara, Italy.,Intrinsic Immunity Unit, CAAD - Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
33
|
Horiguchi H, Loftus TJ, Hawkins RB, Raymond SL, Stortz JA, Hollen MK, Weiss BP, Miller ES, Bihorac A, Larson SD, Mohr AM, Brakenridge SC, Tsujimoto H, Ueno H, Moore FA, Moldawer LL, Efron PA. Innate Immunity in the Persistent Inflammation, Immunosuppression, and Catabolism Syndrome and Its Implications for Therapy. Front Immunol 2018; 9:595. [PMID: 29670613 PMCID: PMC5893931 DOI: 10.3389/fimmu.2018.00595] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/09/2018] [Indexed: 12/12/2022] Open
Abstract
Clinical and technological advances promoting early hemorrhage control and physiologic resuscitation as well as early diagnosis and optimal treatment of sepsis have significantly decreased in-hospital mortality for many critically ill patient populations. However, a substantial proportion of severe trauma and sepsis survivors will develop protracted organ dysfunction termed chronic critical illness (CCI), defined as ≥14 days requiring intensive care unit (ICU) resources with ongoing organ dysfunction. A subset of CCI patients will develop the persistent inflammation, immunosuppression, and catabolism syndrome (PICS), and these individuals are predisposed to a poor quality of life and indolent death. We propose that CCI and PICS after trauma or sepsis are the result of an inappropriate bone marrow response characterized by the generation of dysfunctional myeloid populations at the expense of lympho- and erythropoiesis. This review describes similarities among CCI/PICS phenotypes in sepsis, cancer, and aging and reviews the role of aberrant myelopoiesis in the pathophysiology of CCI and PICS. In addition, we characterize pathogen recognition, the interface between innate and adaptive immune systems, and therapeutic approaches including immune modulators, gut microbiota support, and nutritional and exercise therapy. Finally, we discuss the future of diagnostic and prognostic approaches guided by machine and deep-learning models trained and validated on big data to identify patients for whom these approaches will yield the greatest benefits. A deeper understanding of the pathophysiology of CCI and PICS and continued investigation into novel therapies harbor the potential to improve the current dismal long-term outcomes for critically ill post-injury and post-infection patients.
Collapse
Affiliation(s)
- Hiroyuki Horiguchi
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States.,Department of Surgery, National Defense Medical College, Tokorozawa, Japan
| | - Tyler J Loftus
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Russell B Hawkins
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Steven L Raymond
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Julie A Stortz
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - McKenzie K Hollen
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Brett P Weiss
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Elizabeth S Miller
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Azra Bihorac
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Shawn D Larson
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Alicia M Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Scott C Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, Tokorozawa, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Tokorozawa, Japan
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | | |
Collapse
|
34
|
Rebordão MR, Amaral A, Lukasik K, Szóstek-Mioduchowska A, Pinto-Bravo P, Galvão A, Skarzynski DJ, Ferreira-Dias G. Constituents of neutrophil extracellular traps induce in vitro collagen formation in mare endometrium. Theriogenology 2018; 113:8-18. [PMID: 29452855 DOI: 10.1016/j.theriogenology.2018.02.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 01/08/2023]
Abstract
Neutrophil extracellular traps (NETs) are DNA complexes carrying nuclear and cytoplasmic proteins, such as elastase (ELA), cathepsin-G (CAT) and myeloperoxidase (MPO). Mare endometrosis is a chronic degenerative process characterized by excessive collagen in endometrium. While NETs fight bacteria that cause endometritis, they may trigger endometrial fibrogenesis. The aim was to evaluate the in vitro effect of some NETs components on mare endometrial fibrogenesis and determine its relationship with histopathology or estrous cycle. Endometrial explants were incubated with NETs components (ELA, CAT, MPO or oxytocin). Collagen type I (COL1) protein and type I and III (COL3) gene transcription were evaluated in follicular and mid-luteal phases endometria (Kenney and Doig type I/IIA and IIB/III). Increased COL1 occurred with all NETs proteins, although endometrial response to each NETs protease depended on estrous cycle and/or endometrial category. Since ELA enhanced COL1 production, NETs persistence might be linked to endometrosis. Estrous cycle influenced COL1 protein concentration and COL3 transcripts, suggesting that follicular phase may favor endometrial collagen production. However, luteal phase endometria with moderate or severe lesions may be also susceptible to fibrotic effects of NETs constituents. These data propose that NETs involvement in chronic endometritis in mares may act as putative endometrial fibrogenic mediators.
Collapse
Affiliation(s)
- Maria Rosa Rebordão
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Coimbra College of Agriculture, Polytechnic Institute of Coimbra, Coimbra, Portugal
| | - Ana Amaral
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Karolina Lukasik
- Institute of Animal Reproduction and Food Research of PAS, Olsztyn, Poland
| | | | - Pedro Pinto-Bravo
- Coimbra College of Agriculture, Polytechnic Institute of Coimbra, Coimbra, Portugal
| | - António Galvão
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Institute of Animal Reproduction and Food Research of PAS, Olsztyn, Poland
| | | | - Graça Ferreira-Dias
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
35
|
Beyond the Matrix: The Many Non-ECM Ligands for Integrins. Int J Mol Sci 2018; 19:ijms19020449. [PMID: 29393909 PMCID: PMC5855671 DOI: 10.3390/ijms19020449] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/21/2018] [Accepted: 01/30/2018] [Indexed: 12/17/2022] Open
Abstract
The traditional view of integrins portrays these highly conserved cell surface receptors as mediators of cellular attachment to the extracellular matrix (ECM), and to a lesser degree, as coordinators of leukocyte adhesion to the endothelium. These canonical activities are indispensable; however, there is also a wide variety of integrin functions mediated by non-ECM ligands that transcend the traditional roles of integrins. Some of these unorthodox roles involve cell-cell interactions and are engaged to support immune functions such as leukocyte transmigration, recognition of opsonization factors, and stimulation of neutrophil extracellular traps. Other cell-cell interactions mediated by integrins include hematopoietic stem cell and tumor cell homing to target tissues. Integrins also serve as cell-surface receptors for various growth factors, hormones, and small molecules. Interestingly, integrins have also been exploited by a wide variety of organisms including viruses and bacteria to support infectious activities such as cellular adhesion and/or cellular internalization. Additionally, the disruption of integrin function through the use of soluble integrin ligands is a common strategy adopted by several parasites in order to inhibit blood clotting during hematophagy, or by venomous snakes to kill prey. In this review, we strive to go beyond the matrix and summarize non-ECM ligands that interact with integrins in order to highlight these non-traditional functions of integrins.
Collapse
|
36
|
Regulated Cell Death. DAMAGE-ASSOCIATED MOLECULAR PATTERNS IN HUMAN DISEASES 2018. [PMCID: PMC7123501 DOI: 10.1007/978-3-319-78655-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In this chapter, the various subroutines of regulated cell death are neatly described by highlighting apoptosis and subforms of regulated necrosis such as necroptosis, ferroptosis, pyroptosis, and NETosis. Typically, all forms of regulated necrosis are defined by finite rupture of the plasma cell membrane. Apoptosis is characterized by an enzymatic machinery that consists of caspases which cause the morphologic features of this type of cell death. Mechanistically, apoptosis can be instigated by two major cellular signalling pathways: an intrinsic pathway that is initiated inside cells by mitochondrial release of pro-apoptotic factors or an extrinsic pathway that is initiated at the cell surface by various death receptors. In necroptosis, the biochemical processes are distinct from those found in apoptosis; in particular, there is no caspase activation. As such, necroptosis is a kinase-mediated cell death that relies on “receptor-interacting protein kinase 3” which mediates phosphorylation of the pseudokinase “mixed lineage kinase domain-like protein.” While ferroptosis is an iron-dependent, oxidative form of regulated necrosis that is biochemically characterized by accumulation of ROS from iron metabolism, oxidase activity, and lipid peroxidation products, pyroptosis is defined as a form of cell death (predominantly of phagocytes) that develops during inflammasome activation and is executed by caspase-mediated cleavage of the pore-forming protein gasdermin D. Finally, NETosis refers to a regulated death of neutrophils that is characterized by the release of chromatin-derived weblike structures released into the extracellular space. The chapter ends up with a discussion on the characteristic feature of regulated necrosis: the passive release of large amounts of constitutive DAMPs as a consequence of final plasma membrane rupture as well as the active secretion of inducible DAMPs earlier during the dying process. Notably, per cell death subroutine, the active secretion of inducible DAMPs varies, thereby determining different immunogenicity of dying cells.
Collapse
|
37
|
Neutrophil programming dynamics and its disease relevance. SCIENCE CHINA-LIFE SCIENCES 2017; 60:1168-1177. [PMID: 28971361 DOI: 10.1007/s11427-017-9145-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/21/2017] [Indexed: 12/27/2022]
Abstract
Neutrophils are traditionally considered as first responders to infection and provide antimicrobial host defense. However, recent advances indicate that neutrophils are also critically involved in the modulation of host immune environments by dynamically adopting distinct functional states. Functionally diverse neutrophil subsets are increasingly recognized as critical components mediating host pathophysiology. Despite its emerging significance, molecular mechanisms as well as functional relevance of dynamically programmed neutrophils remain to be better defined. The increasing complexity of neutrophil functions may require integrative studies that address programming dynamics of neutrophils and their pathophysiological relevance. This review aims to provide an update on the emerging topics of neutrophil programming dynamics as well as their functional relevance in diseases.
Collapse
|
38
|
Ali N, Auerbach HE. New-onset acute thrombocytopenia in hospitalized patients: pathophysiology and diagnostic approach. J Community Hosp Intern Med Perspect 2017; 7:157-167. [PMID: 28808508 PMCID: PMC5538216 DOI: 10.1080/20009666.2017.1335156] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 05/22/2017] [Indexed: 12/31/2022] Open
Abstract
Thrombocytopenia is a hematological finding commonly encountered in daily clinical practice from asymptomatic clinic patients to critically ill intensive care unit patients. A broad spectrum of etiologies and variation in clinical presentation often present a diagnostic challenge. Furthermore, concomitant presence of thrombosis and thrombocytopenia, as in cases of thrombotic thrombocytopenia, complicates the management. In hospitalized patients, new-onset thrombocytopenia is an important reason for hematology consultation. Therefore, it is of utmost importance that the etiology is diagnosed accurately. In addition, a basic understanding of the pathophysiology and the differential diagnosis avoids delay in the diagnosis and leads to rapid initiation of treatment. This review will address causes of thrombocytopenia that arises in hospitalized patients with an emphasis on the pathophysiological basis of each disorder.
Collapse
Affiliation(s)
- Naveed Ali
- Department of Internal Medicine, Abington Memorial Hospital / Abington-Jefferson Health, Abington, PA, USA
| | - Herbert E. Auerbach
- Department of Pathology, Abington Memorial Hospital / Abington-Jefferson Health, Abington, PA, USA
| |
Collapse
|