1
|
Marrufo AM, Flores-Mireles AL. Macrophage fate: to kill or not to kill? Infect Immun 2024; 92:e0047623. [PMID: 38829045 PMCID: PMC11385966 DOI: 10.1128/iai.00476-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Macrophages are dynamic innate immune cells that either reside in tissue, serving as sentinels, or recruited as monocytes from bone marrow into inflamed and infected tissue. In response to cues in the tissue microenvironment (TME), macrophages polarize on a continuum toward M1 or M2 with diverse roles in progression and resolution of disease. M1-like macrophages exhibit proinflammatory functions with antimicrobial and anti-tumorigenic activities, while M2-like macrophages have anti-inflammatory functions that generally resolve inflammatory responses and orchestrate a tissue healing process. Given these opposite phenotypes, proper spatiotemporal coordination of macrophage polarization in response to cues within the TME is critical to effectively resolve infectious disease and regulate wound healing. However, if this spatiotemporal coordination becomes disrupted due to persistent infection or dysregulated coagulation, macrophages' inappropriate response to these cues will result in the development of diseases with clinically unfavorable outcomes. Since plasticity and heterogeneity are hallmarks of macrophages, they are attractive targets for therapies to reprogram toward specific phenotypes that could resolve disease and favor clinical prognosis. In this review, we discuss how basic science studies have elucidated macrophage polarization mechanisms in TMEs during infections and inflammation, particularly coagulation. Therefore, understanding the dynamics of macrophage polarization within TMEs in diseases is important in further development of targeted therapies.
Collapse
Affiliation(s)
- Armando M. Marrufo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | | |
Collapse
|
2
|
Jin T, An J, Wu W, Zhou F. Development and Validation of a Machine Learning Model for Bone Metastasis in Prostate Cancer: Based on Inflammatory and Nutritional Indicators. Urology 2024; 190:63-70. [PMID: 38825085 DOI: 10.1016/j.urology.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/20/2024] [Accepted: 05/20/2024] [Indexed: 06/04/2024]
Abstract
OBJECTIVE To establish a predictive model for prostate cancer bone metastasis utilizing multiple machine learning algorithms. METHODS Retrospective analysis of the clinical data of prostate cancer initially diagnosed in the Department of Urology of Gansu Provincial People's Hospital from June 2017 to June 2022. Logistic regression (LR) and least absolute shrinkage and selection operator (LASSO) are used to jointly screen the model features. The filtered features are incorporated into algorithms including LR, random forest (RF), extreme gradient boosting (XGBoost), naive Bayes (NB), k-nearest neighbor (KNN), and decision tree (DT), to develop prostate cancer bone metastasis models. RESULTS A total of 404 patients were finally screened. Gleason score, T stage, N stage, PSA, and ALP were used as features for modeling. The average AUC of the 5-fold cross-validation for each machine learning model in the training set is as follows: LR (AUC=0.9054), RF (AUC=0.9032), NB (AUC=0.8961), KNN (AUC=0.8704), DT (AUC=0.8526), XGBoost (AUC=0.8066). The AUC of each machine learning model in the test set is KNN (AUC=0.9390, 95%CI: 0.8760-1), RF (AUC=0.9290, 95%CI: 0.8718-0.9861), NB (AUC=0.9268, 95%CI: 0.8615-0.9920), LR (AUC=0.9212, 95%CI: 0.8506-0.9917), XGBoost (AUC=0.8292, 95%CI: 0.7442-0.9141), DT (AUC=0.8057, 95%CI: 0.7100-0.9014). A comprehensive evaluation showed that LR performed well in interpretability and clinical applications. CONCLUSION A bone metastasis model of prostate cancer was established, and it was observed that indicators such as inflammation and nutrition had a weak correlation with bone metastasis.
Collapse
Affiliation(s)
- Tongtong Jin
- The First Clinical Medical College, Lanzhou University, Lanzhou; Department of Urology, Gansu Provincial People's Hospital, Lanzhou
| | - Jingjing An
- The First Clinical Medical College, Lanzhou University, Lanzhou
| | - Wangjian Wu
- The First Clinical Medical College, Lanzhou University, Lanzhou; Department of Urology, Gansu Provincial People's Hospital, Lanzhou
| | - Fenghai Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou; Department of Urology, Gansu Provincial People's Hospital, Lanzhou.
| |
Collapse
|
3
|
Xie W, Donat A, Jiang S, Baranowsky A, Keller J. The emerging role of tranexamic acid and its principal target, plasminogen, in skeletal health. Acta Pharm Sin B 2024; 14:2869-2884. [PMID: 39027253 PMCID: PMC11252461 DOI: 10.1016/j.apsb.2024.03.033] [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: 11/22/2023] [Revised: 02/28/2024] [Accepted: 03/14/2024] [Indexed: 07/20/2024] Open
Abstract
The worldwide burden of skeletal diseases such as osteoporosis, degenerative joint disease and impaired fracture healing is steadily increasing. Tranexamic acid (TXA), a plasminogen inhibitor and anti-fibrinolytic agent, is used to reduce bleeding with high effectiveness and safety in major surgical procedures. With its widespread clinical application, the effects of TXA beyond anti-fibrinolysis have been noticed and prompted renewed interest in its use. Some clinical trials have characterized the effects of TXA on reducing postoperative infection rates and regulating immune responses in patients undergoing surgery. Also, several animal studies suggest potential therapeutic effects of TXA on skeletal diseases such as osteoporosis and fracture healing. Although a direct effect of TXA on the differentiation and function of bone cells in vitro was shown, few mechanisms of action have been reported. Here, we summarize recent findings of the effects of TXA on skeletal diseases and discuss the underlying plasminogen-dependent and -independent mechanisms related to bone metabolism and the immune response. We furthermore discuss potential novel indications for TXA application as a treatment strategy for skeletal diseases.
Collapse
Affiliation(s)
- Weixin Xie
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Antonia Donat
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Shan Jiang
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Anke Baranowsky
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| |
Collapse
|
4
|
Bareja A, Lee DE, Ho T, Waitt G, McKay LH, Hannou SA, Orenduff MC, McGreevy KM, Binder A, Ryan CP, Soderblom EJ, Belsky DW, Ferrucci L, Das JK, Banskota N, Kraus VB, Huebner JL, Kraus WE, Huffman KM, Baht GS, Horvath S, Parmer RJ, Miles LA, White JP. Liver-derived plasminogen mediates muscle stem cell expansion during caloric restriction through the plasminogen receptor Plg-R KT. Cell Rep 2024; 43:113881. [PMID: 38442019 PMCID: PMC11075744 DOI: 10.1016/j.celrep.2024.113881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 08/08/2023] [Accepted: 02/13/2024] [Indexed: 03/07/2024] Open
Abstract
An intriguing effect of short-term caloric restriction (CR) is the expansion of certain stem cell populations, including muscle stem cells (satellite cells), which facilitate an accelerated regenerative program after injury. Here, we utilized the MetRSL274G (MetRS) transgenic mouse to identify liver-secreted plasminogen as a candidate for regulating satellite cell expansion during short-term CR. Knockdown of circulating plasminogen prevents satellite cell expansion during short-term CR. Furthermore, loss of the plasminogen receptor KT (Plg-RKT) is also sufficient to prevent CR-related satellite cell expansion, consistent with direct signaling of plasminogen through the plasminogen receptor Plg-RKT/ERK kinase to promote proliferation of satellite cells. Importantly, we are able to replicate many of these findings in human participants from the CALERIE trial. Our results demonstrate that CR enhances liver protein secretion of plasminogen, which signals directly to the muscle satellite cell through Plg-RKT to promote proliferation and subsequent muscle resilience during CR.
Collapse
Affiliation(s)
- Akshay Bareja
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
| | - David E Lee
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
| | - Tricia Ho
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC, USA
| | - Greg Waitt
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC, USA
| | - Lauren H McKay
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA; Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of Chapel Hill, Chapel Hill, NC, USA
| | - Sarah A Hannou
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
| | - Melissa C Orenduff
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
| | - Kristen M McGreevy
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA
| | - Alexandra Binder
- Population Sciences in the Pacific Program (Cancer Epidemiology), University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI 96813, USA; Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA
| | - Calen P Ryan
- Columbia University Mailman School of Public Health, New York, NY, USA
| | - Erik J Soderblom
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC, USA
| | - Daniel W Belsky
- Columbia University Mailman School of Public Health, New York, NY, USA
| | - Luigi Ferrucci
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jayanta Kumar Das
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nirad Banskota
- Longitudinal Studies Section, Translation Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Virginia B Kraus
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA; Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA
| | - Janet L Huebner
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
| | - William E Kraus
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA; Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA
| | - Kim M Huffman
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA; Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA
| | - Gurpreet S Baht
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA; Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA; Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC 27701, USA
| | - Steve Horvath
- Computational Biology and Genomics Core, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA; Altos Labs, San Diego, CA, USA
| | - Robert J Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare System, San Diego, CA, USA; Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Lindsey A Miles
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - James P White
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA; Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA.
| |
Collapse
|
5
|
Kanno Y. The Roles of Fibrinolytic Factors in Bone Destruction Caused by Inflammation. Cells 2024; 13:516. [PMID: 38534360 DOI: 10.3390/cells13060516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, Crohn's disease, periodontitis, and carcinoma metastasis frequently result in bone destruction. Pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, and IL-17 are known to influence bone loss by promoting the differentiation and activation of osteoclasts. Fibrinolytic factors, such as plasminogen (Plg), plasmin, urokinase-type plasminogen activator (uPA), its receptor (uPAR), tissue-type plasminogen activator (tPA), α2-antiplasmin (α2AP), and plasminogen activator inhibitor-1 (PAI-1) are expressed in osteoclasts and osteoblasts and are considered essential in maintaining bone homeostasis by regulating the functions of both osteoclasts and osteoblasts. Additionally, fibrinolytic factors are associated with the regulation of inflammation and the immune system. This review explores the roles of fibrinolytic factors in bone destruction caused by inflammation.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Molecular Pathology, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyotanabe, Kyoto 610-0395, Japan
| |
Collapse
|
6
|
Seillier C, Lesec L, Hélie P, Marie C, Vivien D, Docagne F, Le Mauff B, Toutirais O. Tissue-plasminogen activator effects on the phenotype of splenic myeloid cells in acute inflammation. J Inflamm (Lond) 2024; 21:4. [PMID: 38355547 PMCID: PMC10865617 DOI: 10.1186/s12950-024-00375-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Tissue-plasminogen activator (tPA) is a serine protease well known for its fibrinolytic function. Recent studies indicate that tPA could also modulate inflammation via plasmin generation and/or by receptor mediated signalling in vitro. However, the contribution of tPA in inflammatory processes in vivo has not been fully addressed. Therefore, using tPA-deficient mice, we have analysed the effect of lipopolysaccharide (LPS) challenge on the phenotype of myeloid cells including neutrophils, macrophages and dendritic cells (DCs) in spleen. We found that LPS treatment upregulated the frequency of major histocompatibility class two (MHCII+) macrophages but also, paradoxically, induced a deep downregulation of MHCII molecule level on macrophages and on conventional dendritic cells 2 (cDC2). Expression level of the CD11b integrin, known as a tPA receptor, was upregulated by LPS on MHCII+ macrophages and cDC2, suggesting that tPA effects could be amplified during inflammation. In tPA-/- mice under inflammatory conditions, expression of costimulatory CD86 molecules on MHCII+ macrophages was decreased compared to WT mice, while in steady state the expression of MHCII molecules was higher on macrophages. Finally, we reported that tPA deficiency slightly modified the phenotype of DCs and T cells in acute inflammatory conditions. Overall, our findings indicate that in vivo, LPS injection had an unexpectedly bimodal effect on MHCII expression on macrophages and DCs that consequently might affect adaptive immunity. tPA could also participate in the regulation of the T cell response by modulating the levels of CD86 and MHCII molecules on macrophages.
Collapse
Affiliation(s)
- Célia Seillier
- Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Caen, France
| | - Léonie Lesec
- Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Caen, France
| | - Pauline Hélie
- Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Caen, France
- Present address: Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Charlotte Marie
- Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Caen, France
- UAR 3408-US50 / Centre Universitaire de Ressources Biologiques (CURB), GIP Cyceron, Caen, France
| | - Denis Vivien
- Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Caen, France
- Department of Clinical Research, Caen University Hospital, CHU Caen, France
| | - Fabian Docagne
- Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Caen, France
- Present Address: INSERM, Département de L'information Scientifique Et de La Communication (DISC), 75654, Paris Cedex 13, France
| | - Brigitte Le Mauff
- Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Caen, France
- Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU Caen, France
| | - Olivier Toutirais
- Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Caen, France.
- Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU Caen, France.
| |
Collapse
|
7
|
Miles LA, Bai H, Chakrabarty S, Baik N, Zhang Y, Parmer RJ, Samad F. Overexpression of Plg-R KT protects against adipose dysfunction and dysregulation of glucose homeostasis in diet-induced obese mice. Adipocyte 2023; 12:2252729. [PMID: 37642146 PMCID: PMC10481882 DOI: 10.1080/21623945.2023.2252729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023] Open
Abstract
The plasminogen receptor, Plg-RKT, is a unique cell surface receptor that is broadly expressed in cells and tissues throughout the body. Plg-RKT localizes plasminogen on cell surfaces and promotes its activation to the broad-spectrum serine protease, plasmin. In this study, we show that overexpression of Plg-RKT protects mice from high fat diet (HFD)-induced adipose and metabolic dysfunction. During the first 10 weeks on the HFD, the body weights of mice that overexpressed Plg-RKT (Plg-RKT-OEX) were lower than those of control mice (CagRosaPlgRKT). After 10 weeks on the HFD, CagRosaPlgRKT and Plg-RKT-OEX mice had similar body weights. However, Plg-RKT-OEX mice showed a more metabolically favourable body composition phenotype. Plg-RKT-OEX mice also showed improved glucose tolerance and increased insulin sensitivity. We found that the improved metabolic functions of Plg-RKT-OEX mice were mechanistically associated with increased energy expenditure and activity, decreased proinflammatory adipose macrophages and decreased inflammation, elevated brown fat thermogenesis, and higher expression of adipose PPARγ and adiponectin. These findings suggest that Plg-RKT signalling promotes healthy adipose function via multiple mechanisms to defend against obesity-associated adverse metabolic phenotypes.
Collapse
Affiliation(s)
- Lindsey A. Miles
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | - Hongdong Bai
- Department of Medicine, Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Sagarika Chakrabarty
- Department of Cell Biology, San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Nagyung Baik
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | - Yuqing Zhang
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare System, San Diego, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Fahumiya Samad
- Department of Cell Biology, San Diego Biomedical Research Institute, San Diego, CA, USA
| |
Collapse
|
8
|
Xu A, Yang Y, Shao Y, Jiang M, Sun Y, Feng B. FHL2 regulates microglia M1/M2 polarization after spinal cord injury via PARP14-depended STAT1/6 pathway. Int Immunopharmacol 2023; 124:110853. [PMID: 37708708 DOI: 10.1016/j.intimp.2023.110853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
Neuronal apoptosis and inflammation exacerbate the secondary injury after spinal cord injury (SCI). Four and a half domains 2 (FHL2) is a multifunctional scaffold protein with tissue- and cell-type specific effects on the regulation of inflammation, but its role in SCI remains unclear. The T10 mouse spinal cord contusion model was established, and the mice were immediately injected with lentiviruses carrying FHL2 shRNA after SCI. The results showed that FHL2 expression was increased following SCI, and then gradually decreased. Moreover, FHL2 depletion aggravated functional impairment, neuronal necrosis, and enlarged lesion cavity areas in the injured spinal cord. FHL2 deficiency facilitated neuronal apoptosis by elevating cleaved caspase 3/9 expression, neuroinflammation by regulating microglia polarization, and bone loss. Indeed, FHL2 deficiency increased the secretion of TNF-α and IL-6, M1 microglia polarization, and the activation of STAT1 pathway but decreased the secretion of IL-10 and IL-4, M2 microglia polarization, and the activation of the STAT6 pathway in the spinal cord. In vitro, FHL2 silencing promoted LPS + IFN-γ-induced microglia M1 polarization through activating the STAT1 pathway and alleviated IL-4-induced microglia M2 polarization via inhibiting the STAT6 pathway. FHL2 positively regulated the expression of poly (ADP-ribose) polymerase family member 14 (PARP14) by promoting its transcription. PARP14 overexpression inhibited FHL2 silencing-induced microglia M1 polarization and relieved the inhibitory effect of FHL2 silencing on microglia M2 polarization. Collectively, the study suggests that FHL2 reduces the microglia M1/M2 polarization-mediated inflammation via PARP14-dependent STAT1/6 pathway and thereby improves functional recovery after SCI.
Collapse
Affiliation(s)
- Aihua Xu
- Department of Rehabilitation Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Yang
- Department of Rehabilitation Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Shao
- Department of Rehabilitation Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Manyu Jiang
- Department of Rehabilitation Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yongxin Sun
- Department of Rehabilitation Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Bo Feng
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
9
|
Siddiqui H, Deo N, Rutledge MT, Williams MJ, Redpath GM, McCormick SP. Plasminogen Receptors Promote Lipoprotein(a) Uptake by Enhancing Surface Binding and Facilitating Macropinocytosis. Arterioscler Thromb Vasc Biol 2023; 43:1851-1866. [PMID: 37589135 PMCID: PMC10521804 DOI: 10.1161/atvbaha.123.319344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND High levels of Lp(a) (lipoprotein(a)) are associated with multiple forms of cardiovascular disease. Lp(a) consists of an apoB100-containing particle attached to the plasminogen homologue apo(a). The pathways for Lp(a) clearance are not well understood. We previously discovered that the plasminogen receptor PlgRKT (plasminogen receptor with a C-terminal lysine) promoted Lp(a) uptake in liver cells. Here, we aimed to further define the role of PlgRKT and to investigate the role of 2 other plasminogen receptors, annexin A2 and S100A10 (S100 calcium-binding protein A10) in the endocytosis of Lp(a). METHODS Human hepatocellular carcinoma (HepG2) cells and haploid human fibroblast-like (HAP1) cells were used for overexpression and knockout of plasminogen receptors. The uptake of Lp(a), LDL (low-density lipoprotein), apo(a), and endocytic cargos was visualized and quantified by confocal microscopy and Western blotting. RESULTS The uptake of both Lp(a) and apo(a), but not LDL, was significantly increased in HepG2 and HAP1 cells overexpressing PlgRKT, annexin A2, or S100A10. Conversely, Lp(a) and apo(a), but not LDL, uptake was significantly reduced in HAP1 cells in which PlgRKT and S100A10 were knocked out. Surface binding studies in HepG2 cells showed that overexpression of PlgRKT, but not annexin A2 or S100A10, increased Lp(a) and apo(a) plasma membrane binding. Annexin A2 and S100A10, on the other hand, appeared to regulate macropinocytosis with both proteins significantly increasing the uptake of the macropinocytosis marker dextran when overexpressed in HepG2 and HAP1 cells and knockout of S100A10 significantly reducing dextran uptake. Bringing these observations together, we tested the effect of a PI3K (phosphoinositide-3-kinase) inhibitor, known to inhibit macropinocytosis, on Lp(a) uptake. Results showed a concentration-dependent reduction confirming that Lp(a) uptake was indeed mediated by macropinocytosis. CONCLUSIONS These findings uncover a novel pathway for Lp(a) endocytosis involving multiple plasminogen receptors that enhance surface binding and stimulate macropinocytosis of Lp(a). Although the findings were produced in cell culture models that have limitations, they could have clinical relevance since drugs that inhibit macropinocytosis are in clinical use, that is, the PI3K inhibitors for cancer therapy and some antidepressant compounds.
Collapse
Affiliation(s)
- Halima Siddiqui
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Nikita Deo
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Malcolm T. Rutledge
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Michael J.A. Williams
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- Department of Medicine (M.J.A.W.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Gregory M.I. Redpath
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Sally P.A. McCormick
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| |
Collapse
|
10
|
Perucci LO, Vago JP, Miles LA, Sousa LP. Crosstalk between the plasminogen/plasmin system and inflammation resolution. J Thromb Haemost 2023; 21:2666-2678. [PMID: 37495082 PMCID: PMC10792525 DOI: 10.1016/j.jtha.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/29/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
The plasminogen/plasmin (Plg/Pla) system, best known for its classical role in thrombolysis, has been recently highlighted as a regulator of other biological processes in mammals, including key steps involved in the resolution of inflammation. Inflammation resolution is a complex process coordinated by different cellular effectors, notably leukocytes, and active mediators, and is initiated shortly after the inflammatory response begins. Once the inflammatory insult is eliminated, an effective and timely engagement of proresolution programs prevents persistent inflammation, thereby avoiding excessive tissue damage, fibrosis, and the development of autoimmunity. Interestingly, recent studies demonstrate that Plg/Pla and their receptor, plasminogen receptor KT (Plg-RKT), regulate key steps in inflammation resolution. The number of studies investigating the involvement of the Plg/Pla system in these and other aspects of inflammation, including degradation of extracellular matrices, immune cell migration, wound healing, and skeletal growth and maintenance, highlights key roles of the Plg/Pla system during physiological and pathologic conditions. Here, we discuss robust evidence in the literature for the emerging roles of the Plg/Pla system in key steps of inflammation resolution. These findings suggest that dysregulation in Plg production and its activation plays a role in the pathogenesis of inflammatory diseases. Elucidating central mechanisms underlying the role of Plg/Pla in key steps of inflammation resolution either in preclinical models of inflammation or in human inflammatory conditions, can provide a rationale for the development of new pharmacologic interventions to promote resolution of inflammation, and open new pathways for the treatment of thromboinflammatory conditions.
Collapse
Affiliation(s)
- Luiza O Perucci
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Juliana P Vago
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lindsey A Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Lirlândia P Sousa
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
11
|
Satala D, Bednarek A, Kozik A, Rapala-Kozik M, Karkowska-Kuleta J. The Recruitment and Activation of Plasminogen by Bacteria-The Involvement in Chronic Infection Development. Int J Mol Sci 2023; 24:10436. [PMID: 37445613 DOI: 10.3390/ijms241310436] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
The development of infections caused by pathogenic bacteria is largely related to the specific properties of the bacterial cell surface and extracellular hydrolytic activity. Furthermore, a significant role of hijacking of host proteolytic cascades by pathogens during invasion should not be disregarded during consideration of the mechanisms of bacterial virulence. This is the key factor for the pathogen evasion of the host immune response, tissue damage, and pathogen invasiveness at secondary infection sites after initial penetration through tissue barriers. In this review, the mechanisms of bacterial impact on host plasminogen-the precursor of the important plasma serine proteinase, plasmin-are characterized, principally focusing on cell surface exposition of various proteins, responsible for binding of this host (pro)enzyme and its activators or inhibitors, as well as the fibrinolytic system activation tactics exploited by different bacterial species, not only pathogenic, but also selected harmless residents of the human microbiome. Additionally, the involvement of bacterial factors that modulate the process of plasminogen activation and fibrinolysis during periodontitis is also described, providing a remarkable example of a dual use of this host system in the development of chronic diseases.
Collapse
Affiliation(s)
- Dorota Satala
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Aneta Bednarek
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-387 Kraków, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| |
Collapse
|
12
|
Whyte CS. All tangled up: interactions of the fibrinolytic and innate immune systems. Front Med (Lausanne) 2023; 10:1212201. [PMID: 37332750 PMCID: PMC10272372 DOI: 10.3389/fmed.2023.1212201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
The hemostatic and innate immune system are intertwined processes. Inflammation within the vasculature promotes thrombus development, whilst fibrin forms part of the innate immune response to trap invading pathogens. The awareness of these interlinked process has resulted in the coining of the terms "thromboinflammation" and "immunothrombosis." Once a thrombus is formed it is up to the fibrinolytic system to resolve these clots and remove them from the vasculature. Immune cells contain an arsenal of fibrinolytic regulators and plasmin, the central fibrinolytic enzyme. The fibrinolytic proteins in turn have diverse roles in immunoregulation. Here, the intricate relationship between the fibrinolytic and innate immune system will be discussed.
Collapse
|
13
|
Vizzoni L, Migone C, Grassiri B, Zambito Y, Ferro B, Roncucci P, Mori F, Salvatore A, Ascione E, Crea R, Esin S, Batoni G, Piras AM. Biopharmaceutical Assessment of Mesh Aerosolised Plasminogen, a Step towards ARDS Treatment. Pharmaceutics 2023; 15:1618. [PMID: 37376068 PMCID: PMC10300680 DOI: 10.3390/pharmaceutics15061618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe complication of lung injuries, commonly associated with bacterial, fungal and viral infections, including SARS-CoV-2 viral infections. ARDS is strongly correlated with patient mortality and its clinical management is very complex, with no effective treatment presently available. ARDS involves severe respiratory failure, fibrin deposition in both airways and lung parenchyma, with the development of an obstructing hyaline membrane drastically limiting gas exchange. Moreover, hypercoagulation is related to deep lung inflammation, and a pharmacological action toward both aspects is expected to be beneficial. Plasminogen (PLG) is a main component of the fibrinolytic system playing key roles in various inflammation regulatory processes. The inhalation of PLG has been proposed in the form of the off-label administration of an eyedrop solution, namely, a plasminogen-based orphan medicinal product (PLG-OMP), by means of jet nebulisation. Being a protein, PLG is susceptible to partial inactivation under jet nebulisation. The aim of the present work is to demonstrate the efficacy of the mesh nebulisation of PLG-OMP in an in vitro simulation of clinical off-label administration, considering both the enzymatic and immunomodulating activities of PLG. Biopharmaceutical aspects are also investigated to corroborate the feasibility of PLG-OMP administration by inhalation. The nebulisation of the solution was performed using an Aerogen® SoloTM vibrating-mesh nebuliser. Aerosolised PLG showed an optimal in vitro deposition profile, with 90% of the active ingredient impacting the lower portions of a glass impinger. The nebulised PLG remained in its monomeric form, with no alteration of glycoform composition and 94% of enzymatic activity maintenance. Activity loss was observed only when PLG-OMP nebulisation was performed under simulated clinical oxygen administration. In vitro investigations evidenced good penetration of aerosolised PLG through artificial airway mucus, as well as poor permeation across an Air-Liquid Interface model of pulmonary epithelium. The results suggest a good safety profile of inhalable PLG, excluding high systemic absorption but with good mucus diffusion. Most importantly, the aerosolised PLG was capable of reversing the effects of an LPS-activated macrophage RAW 264.7 cell line, demonstrating the immunomodulating activity of PLG in an already induced inflammatory state. All physical, biochemical and biopharmaceutical assessments of mesh aerosolised PLG-OMP provided evidence for its potential off-label administration as a treatment for ARDS patients.
Collapse
Affiliation(s)
- Lucia Vizzoni
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Chiara Migone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Ylenia Zambito
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Research Centre for Nutraceutical and Healthy Foods “NUTRAFOOD”, University of Pisa, 56124 Pisa, Italy
| | - Baldassare Ferro
- Anestesia e Rianimazione, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy
| | - Paolo Roncucci
- Anestesia e Rianimazione, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy
| | - Filippo Mori
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Alfonso Salvatore
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Ester Ascione
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Roberto Crea
- Kedrion S.p.A., Via di Fondovalle, Loc. Bolognana, 55027 Gallicano, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| | - Anna Maria Piras
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Centre for Instrument Sharing of University of Pisa (CISUP), 56126 Pisa, Italy
| |
Collapse
|
14
|
Grossi LC, Zaidan I, Souza JAM, Carvalho AFS, Sanches RCO, Cardoso C, Lara ES, Montuori-Andrade ACM, Bruscoli S, Marchetti MC, Riccardi C, Teixeira MM, Tavares LP, Vago JP, Sousa LP. GILZ Modulates the Recruitment of Monocytes/Macrophages Endowed with a Resolving Phenotype and Favors Resolution of Escherichia coli Infection. Cells 2023; 12:1403. [PMID: 37408237 DOI: 10.3390/cells12101403] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 04/30/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
Macrophages are important effectors of inflammation resolution that contribute to the elimination of pathogens and apoptotic cells and restoration of homeostasis. Pre-clinical studies have evidenced the anti-inflammatory and pro-resolving actions of GILZ (glucocorticoid-induced leucine zipper). Here, we evaluated the role of GILZ on the migration of mononuclear cells under nonphlogistic conditions and Escherichia coli-evoked peritonitis. TAT-GILZ (a cell-permeable GILZ-fusion protein) injection into the pleural cavity of mice induced monocyte/macrophage influx alongside increased CCL2, IL-10 and TGF-β levels. TAT-GILZ-recruited macrophages showed a regulatory phenotype, exhibiting increased expression of CD206 and YM1. During the resolving phase of E. coli-induced peritonitis, marked by an increased recruitment of mononuclear cells, lower numbers of these cells and CCL2 levels were found in the peritoneal cavity of GILZ-deficient mice (GILZ-/-) when compared to WT. In addition, GILZ-/- showed higher bacterial loads, lower apoptosis/efferocytosis counts and a lower number of macrophages with pro-resolving phenotypes. TAT-GILZ accelerated resolution of E. coli-evoked neutrophilic inflammation, which was associated with increased peritoneal numbers of monocytes/macrophages, enhanced apoptosis/efferocytosis counts and bacterial clearance through phagocytosis. Taken together, we provided evidence that GILZ modulates macrophage migration with a regulatory phenotype, inducing bacterial clearance and accelerating the resolution of peritonitis induced by E. coli.
Collapse
Affiliation(s)
- Laís C Grossi
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Isabella Zaidan
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Jéssica Amanda Marques Souza
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Antônio Felipe S Carvalho
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Hospital das Clínicas da Universidade Federal de Minas Gerais/Ebserh, Belo Horizonte 30130-100, Brazil
| | - Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Camila Cardoso
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Edvaldo S Lara
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Ana Clara M Montuori-Andrade
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Stefano Bruscoli
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy
| | - Maria Cristina Marchetti
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy
| | - Carlo Riccardi
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy
| | - Mauro M Teixeira
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Luciana P Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Juliana P Vago
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Lirlândia P Sousa
- Signaling in Inflammation Lab., Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| |
Collapse
|
15
|
Yang Y, Li W, Liu C, Liu J, Yang L, Yue W, Yang L, Xue R, Zhang K, Zhang H, Chang N, Li L. Single-cell RNA seq identifies Plg-R KT-PLG as signals inducing phenotypic transformation of scar-associated macrophage in liver fibrosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166754. [PMID: 37207518 DOI: 10.1016/j.bbadis.2023.166754] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/21/2023]
Abstract
Hepatic macrophages play a central role in liver fibrosis. Scar-associated macrophages (SAMs), a recently identified subgroup of macrophages, play an important role in this process. However, the mechanism by which SAMs transform during liver fibrosis is still unclear. In this study, we aimed to characterize SAMs and elucidate the underlying mechanism of SAM transformation. Bile duct ligation (BDL) and carbon tetrachloride (CCl4) were used to induce mouse liver fibrosis. Non-parenchymal cells were isolated from normal/fibrotic livers and were analyzed using single cell RNA sequencing (scRNA-seq) or mass cytometry (CyTOF). The glucan-encapsulated siRNA particles (siRNA-GeRPs) was employed to perform macrophage selective gene knockdown. The results of scRNA-seq and CyTOF revealed that SAMs, which derived from bone marrow-derived macrophages (BMMs), accumulated in mouse fibrotic livers. Further analysis showed that SAMs highly expressed genes related to fibrosis, indicating the pro-fibrotic functions of SAMs. Moreover, plasminogen receptor Plg-RKT was highly expressed by SAMs, suggesting the role of Plg-RKT and plasminogen (PLG) in SAM transformation. In vitro, PLG-treated BMMs transformed into SAMs and expressed SAM functional genes. Knockdown of Plg-RKT blocked the effects of PLG. In vivo, selective knockdown of Plg-RKT in intrahepatic macrophages of BDL- and CCl4-treated mice reduced the number of SAMs and alleviated BDL- and CCl4-induced liver fibrosis, suggesting that Plg-RKT-PLG played an important role in liver fibrosis by mediating SAM transformation. Our findings reveal that SAMs are crucial participants in liver fibrosis. Inhibition of SAM transformation by blocking Plg-RKT might be a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Yuanru Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Weiyang Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Chang Liu
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Jing Liu
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Lin Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Wenhui Yue
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Le Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Renmin Xue
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Kai Zhang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Hang Zhang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China
| | - Na Chang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China.
| | - Liying Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
16
|
Vago JP, Zaidan I, Perucci LO, Brito LF, Teixeira LC, Silva CMS, Miranda TC, Melo EM, Bruno AS, Queiroz-Junior CM, Sugimoto MA, Tavares LP, Grossi LC, Borges IN, Schneider AH, Baik N, Schneider AH, Talvani A, Ferreira RG, Alves-Filho JC, Nobre V, Teixeira MM, Parmer RJ, Miles LA, Sousa LP. Plasmin and plasminogen prevent sepsis severity by reducing neutrophil extracellular traps and systemic inflammation. JCI Insight 2023; 8:e166044. [PMID: 36917195 PMCID: PMC10243804 DOI: 10.1172/jci.insight.166044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Sepsis is a lethal syndrome characterized by systemic inflammation and abnormal coagulation. Despite therapeutic advances, sepsis mortality remains substantially high. Herein, we investigated the role of the plasminogen/plasmin (Plg/Pla) system during sepsis. Plasma levels of Plg were significantly lower in mice subjected to severe compared with nonsevere sepsis, whereas systemic levels of IL-6, a marker of sepsis severity, were higher in severe sepsis. Plg levels correlated negatively with IL-6 in both septic mice and patients, whereas plasminogen activator inhibitor-1 levels correlated positively with IL-6. Plg deficiency render mice susceptible to nonsevere sepsis induced by cecal ligation and puncture (CLP), resulting in greater numbers of neutrophils and M1 macrophages, liver fibrin(ogen) deposition, lower efferocytosis, and increased IL-6 and neutrophil extracellular trap (NET) release associated with organ damage. Conversely, inflammatory features, fibrin(ogen), and organ damage were substantially reduced, and efferocytosis was increased by exogenous Pla given during CLP- and LPS-induced endotoxemia. Plg or Pla protected mice from sepsis-induced lethality and enhanced the protective effect of antibiotics. Mechanistically, Plg/Pla-afforded protection was associated with regulation of NET release, requiring Pla-protease activity and lysine binding sites. Plg/Pla are important host-protective players during sepsis, controlling local and systemic inflammation and collateral organ damage.
Collapse
Affiliation(s)
- Juliana P. Vago
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Isabella Zaidan
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Luiza O. Perucci
- Department of Biological Sciences, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Larissa Froede Brito
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Lívia C.R. Teixeira
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Camila Meirelles Souza Silva
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Thaís C. Miranda
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Eliza M. Melo
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alexandre S. Bruno
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michelle A. Sugimoto
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana P. Tavares
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laís C. Grossi
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Isabela N. Borges
- Hospital of Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ayda Henriques Schneider
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Nagyung Baik
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Ayda H. Schneider
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - André Talvani
- Department of Biological Sciences, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Raphael G. Ferreira
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - José C. Alves-Filho
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Vandack Nobre
- Hospital of Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M. Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare System and University of California, San Diego, California, USA
| | - Lindsey A. Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Lirlândia P. Sousa
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| |
Collapse
|
17
|
Rocha MP, Oliveira DP, de Oliveira VLS, Zaidan I, Grossi LC, Campana PRV, Amaral FA, Sousa LP, Teixeira MM, Braga FC. Ouratea spectabilis and its Biflavanone Ouratein D Exert Potent Anti-inflammatory Activity in MSU Crystal-induced Gout in Mice. PLANTA MEDICA 2023. [PMID: 36626932 DOI: 10.1055/a-2009-9809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Gouty arthritis (GA) is an inflammatory arthritis triggered by the deposition of monosodium urate monohydrate (MSU) crystals, causing pain, inflammation, and joint damage. Several drugs are currently employed to manage acute flares of GA, but they either have limited effectiveness or induce severe adverse reactions. Ouratea spectabilis is traditionally used in Brazil to treat gastric ulcers and rheumatism. The ethanolic extract of O. spectabilis stems (OSpC) and four biflavanones (ouratein A - D) isolated thereof were evaluated in a murine model of GA induced by the injection of MSU crystals. The underlying mechanism of action of ouratein D was investigated in vitro in cell cultures by measurement of IL-1β levels by ELISA and Western blot analysis. The administration of OSpC (10, 30 or 100 mg/Kg, p. o.) reduced the migration of total inflammatory cells, monocytes, and neutrophils and diminished the levels of IL-1β and CXCL1 in the synovial tissue. Among the tested compounds, only ouratein D (1 mg/Kg) reduced the migration of the inflammatory cells and it was shown to be active up to 0.01 mg/Kg (equivalent to 0.34 nM/Kg, p. o.). Treatment of pre-stimulated THP-1 cells (differentiated into macrophages) or BMDMs with ouratein D reduced the release of IL-1β in both macrophage lines. This biflavanone reduced the activation of caspase-1 (showed by the increase in the cleaved form) in supernatants of cultured BMDMs, evidencing its action in modulating the inflammasome pathway. The obtained results demonstrate the anti-gout properties of O. spectabilis and point out ouratein D as the bioactive component of the assayed extract.
Collapse
Affiliation(s)
- Marina P Rocha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Diego P Oliveira
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian L S de Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Zaidan
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laís C Grossi
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Priscilla R V Campana
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio A Amaral
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernão C Braga
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
18
|
The uPA/uPAR System Orchestrates the Inflammatory Response, Vascular Homeostasis, and Immune System in Fibrosis Progression. Int J Mol Sci 2023; 24:ijms24021796. [PMID: 36675310 PMCID: PMC9866279 DOI: 10.3390/ijms24021796] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Fibrotic diseases, such as systemic sclerosis (SSc), idiopathic pulmonary fibrosis, renal fibrosis and liver cirrhosis are characterized by tissue overgrowth due to excessive extracellular matrix (ECM) deposition. Fibrosis progression is caused by ECM overproduction and the inhibition of ECM degradation due to several events, including inflammation, vascular endothelial dysfunction, and immune abnormalities. Recently, it has been reported that urokinase plasminogen activator (uPA) and its receptor (uPAR), known to be fibrinolytic factors, orchestrate the inflammatory response, vascular homeostasis, and immune homeostasis system. The uPA/uPAR system may show promise as a potential therapeutic target for fibrotic diseases. This review considers the role of the uPA/uPAR system in the progression of fibrotic diseases.
Collapse
|
19
|
Al-Kuraishy HM, Al-Gareeb AI, Al-Harcan NAH, Alexiou A, Batiha GES. Tranexamic Acid and Plasminogen/Plasmin Glaring Paradox in COVID-19. Endocr Metab Immune Disord Drug Targets 2023; 23:35-45. [PMID: 35927893 DOI: 10.2174/1871530322666220801102402] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 11/22/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a severe acute respiratory syndrome, coronavirus type 2 (SARS-CoV-2), leading to acute tissue injury and an overstated immune response. In COVID-19, there are noteworthy changes in the fibrinolytic system with the development of coagulopathy. Therefore, modulation of the fibrinolytic system may affect the course of COVID-19. Tranexamic acid (TXA) is an anti-fibrinolytic drug that reduces the conversion of plasminogen to plasmin, which is necessary for SARS-CoV-2 infectivity. In addition, TXA has anti-inflammatory, anti-platelet, and anti-thrombotic effects, which may attenuate the COVID-19 severity. Thus, in this narrative review, we try to find the beneficial and harmful effects of TXA in COVID-19.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyiah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyiah University, Baghdad, Iraq
| | - Nasser A Hadi Al-Harcan
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Rasheed University College, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia.,AFNP Med Austria, Wien, Austria
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| |
Collapse
|
20
|
Sugimoto MA, Perucci LO, Tavares LP, Teixeira MM, Sousa LP. Fibrinolysis in COVID-19: Impact on Clot Lysis and Modulation of Inflammation. Curr Drug Targets 2022; 23:1578-1592. [PMID: 36221881 DOI: 10.2174/1389450123666221011102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/26/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023]
Abstract
COVID-19 is a multisystem disease caused by SARS-CoV-2 and is associated with an imbalance between the coagulation and fibrinolytic systems. Overall, hypercoagulation, hypofibrinolysis and fibrin-clot resistance to fibrinolysis predispose patients to thrombotic and thromboembolic events. In the lungs, the virus triggers alveolar and interstitial fibrin deposition, endothelial dysfunction, and pulmonary intravascular coagulation, all events intrinsically associated with the activation of inflammation and organ injury. Adding to the pathogenesis of COVID-19, there is a positive feedback loop by which local fibrin deposition in the lungs can fuel inflammation and consequently dysregulates coagulation, a process known as immunothrombosis. Therefore, fibrinolysis plays a central role in maintaining hemostasis and tissue homeostasis during COVID-19 by cleaning fibrin clots and controlling feed-forward products of coagulation. In addition, components of the fibrinolytic system have important immunomodulatory roles, as evidenced by studies showing the contribution of Plasminogen/Plasmin (Plg/Pla) to the resolution of inflammation. Herein, we review clinical evidence for the dysregulation of the fibrinolytic system and discuss its contribution to thrombosis risk and exacerbated inflammation in severe COVID-19. We also discuss the current concept of an interplay between fibrinolysis and inflammation resolution, mirroring the well-known crosstalk between inflammation and coagulation. Finally, we consider the central role of the Plg/Pla system in resolving thromboinflammation, drawing attention to the overlooked consequences of COVID-19-associated fibrinolytic abnormalities to local and systemic inflammation.
Collapse
Affiliation(s)
- Michelle A Sugimoto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Division of Medicine, University College London, London, UK.,Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luiza O Perucci
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Nucleus of Research on Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Luciana P Tavares
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Lirlândia P Sousa
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
21
|
Hu YH, Wang XY, Zhang XW, Chen J, Li F. Investigation of the mechanisms and experimental verification of Shao yao gan cao decoction against Sphincter of Oddi Dysfunction via systems pharmacology. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:13374-13398. [PMID: 36654051 DOI: 10.3934/mbe.2022626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
This study explored the chemical and pharmacological mechanisms of Shao Yao Gan Cao decoction (SYGC) in the treatment of Sphincter of Oddi Dysfunction (SOD) through ultra-high-performance liquid chromatography coupled with Quadrupole Exactive-Orbitrap high-resolution mass spectrometry (UHPLC-Q Exactive-Orbitrap HR-MS), network pharmacology, transcriptomics, molecular docking and in vivo experiments. First, we identified that SYGC improves SOD in guinea pigs by increased c-kit expression and decreased inflammation infiltration and ring muscle disorders. Then, a total of 649 SOD differential genes were found through RNA sequencing and mainly enriched in complement and coagulation cascades, the B cell receptor signaling pathway and the NF-kappa B signaling pathway. By combining UHPLC-Q-Orbitrap-HRMS with a network pharmacology study, 111 chemicals and a total of 52 common targets were obtained from SYGC in the treatment of SOD, which is also involved in muscle contraction, the B cell receptor signaling pathway and the complement system. Next, 20 intersecting genes were obtained among the PPI network, MCODE and ClusterOne analysis. Then, the molecular docking results indicated that four active compounds (glycycoumarin, licoflavonol, echinatin and homobutein) and three targets (AURKB, KIF11 and PLG) exerted good binding interactions, which are also related to the B cell receptor signaling pathway and the complement system. Finally, animal experiments were conducted to confirm the SYGC therapy effects on SOD and verify the 22 hub genes using RT-qPCR. This study demonstrates that SYGC confers therapeutic effects against an experimental model of SOD via regulating immune response and inflammation, which provides a basis for future research and clinical applications.
Collapse
Affiliation(s)
- Yong-Hong Hu
- Institute of Digestive Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xue-Ying Wang
- Department of Preventive Treatment, Shuguang Hospital affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xi-Wen Zhang
- Department of Pancreaticobiliary Surgery, Shuguang Hospital affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jian Chen
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
- Institute of Vascular Anomalies, Shanghai Academy of Traditional Chinese Medicine, Shanghai 200082, China
| | - Fu Li
- Department of Pancreaticobiliary Surgery, Shuguang Hospital affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
22
|
Ariëns RA, Hunt BJ, Agbani EO, Ahnström J, Ahrends R, Alikhan R, Assinger A, Bagoly Z, Balduini A, Barbon E, Barrett CD, Batty P, Carneiro JDA, Chan W, de Maat M, de Wit K, Denis C, Ellis MH, Eslick R, Fu H, Hayward CPM, Ho‐Tin‐Noé B, Klok F, Kumar R, Leiderman K, Litvinov RI, Mackman N, McQuilten Z, Neal MD, Parker WAE, Preston RJS, Rayes J, Rezaie AR, Roberts LN, Rocca B, Shapiro S, Siegal DM, Sousa LP, Suzuki‐Inoue K, Zafar T, Zhou J. Illustrated State-of-the-Art Capsules of the ISTH 2022 Congress. Res Pract Thromb Haemost 2022; 6:e12747. [PMID: 35814801 PMCID: PMC9257378 DOI: 10.1002/rth2.12747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
The ISTH London 2022 Congress is the first held (mostly) face-to-face again since the COVID-19 pandemic took the world by surprise in 2020. For 2 years we met virtually, but this year's in-person format will allow the ever-so-important and quintessential creativity and networking to flow again. What a pleasure and joy to be able to see everyone! Importantly, all conference proceedings are also streamed (and available recorded) online for those unable to travel on this occasion. This ensures no one misses out. The 2022 scientific program highlights new developments in hemophilia and its treatment, acquired and other inherited bleeding disorders, thromboinflammation, platelets and coagulation, clot structure and composition, fibrinolysis, vascular biology, venous thromboembolism, women's health, arterial thrombosis, pediatrics, COVID-related thrombosis, vaccine-induced thrombocytopenia with thrombosis, and omics and diagnostics. These areas are elegantly reviewed in this Illustrated Review article. The Illustrated Review is a highlight of the ISTH Congress. The format lends itself very well to explaining the science, and the collection of beautiful graphical summaries of recent developments in the field are stunning and self-explanatory. This clever and effective way to communicate research is revolutionary and different from traditional formats. We hope you enjoy this article and will be inspired by its content to generate new research ideas.
Collapse
Affiliation(s)
| | | | - Ejaife O. Agbani
- Department of Physiology and Pharmacology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | | | - Robert Ahrends
- Institute of Analytical ChemistryUniversity of ViennaViennaAustria
| | - Raza Alikhan
- Haemostasis & ThrombosisUniversity Hospital of WalesCardiffUK
| | | | - Zsuzsa Bagoly
- Faculty of Medicine, Department of Laboratory Medicine, Division of Clinical Laboratory Sciences and ELKH‐DE Neurodegenerative and Cerebrovascular Research GroupUniversity of DebrecenDebrecenHungary
| | | | - Elena Barbon
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Christopher D. Barrett
- Division of Acute Care Surgery and Surgical Critical Care, Department of SurgeryUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Koch Institute, Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Division of Surgical Critical Care, Department of Surgery, Boston University Medical CenterBoston University School of MedicineBostonMassachusettsUSA
| | | | | | - Wee Shian Chan
- University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Moniek de Maat
- Department of HematologyErasmus MCRotterdamThe Netherlands
| | - Kerstin de Wit
- Queen’s University and McMaster UniversityKingstonONCanada
| | | | - Martin H. Ellis
- Hematology Institute and Blood Bank, Meir Medical Center and Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Renee Eslick
- Haematology DepartmentCanberra HospitalGarranAustralian Capital TerritoryAustralia
| | - Hongxia Fu
- Division of Hematology, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | | | | | - Frederikus A. Klok
- Department of Medicine – Thrombosis and HemostasisLeiden University Medical CenterLeidenThe Netherlands
| | - Riten Kumar
- Dana Farber/Boston Children’s Cancer and Blood Disorders CenterBostonMassachusettsUSA
| | | | - Rustem I. Litvinov
- Department of Cell and Developmental BiologyUniversity of Pennsylvania School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | | | - Matthew D. Neal
- Trauma and Transfusion Medicine Research Center, Department of SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - William A. E. Parker
- Cardiovascular Research Unit, Northern General HospitalUniversity of SheffieldSheffieldUK
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, Department of Pharmacy & Biomolecular SciencesRoyal College of Surgeons in IrelandDublin 2Ireland
| | | | - Alireza R. Rezaie
- Cardiovascular Biology Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Lara N. Roberts
- King’s Thrombosis Centre, Department of Haematological MedicineKing’s College Hospital NHS Foundation TrustLondonUK
| | - Bianca Rocca
- Department of Safety and Bioethics, Section of PharmacologyCatholic University School of MedicineRomeItaly
| | - Susan Shapiro
- Oxford University Hospitals NHS Foundation TrustOxfordUK
- Radcliffe Department of MedicineOxford UniversityOxfordUK
| | - Deborah M. Siegal
- Ottawa Hospital Research Institute and University of OttawaOttawaOntarioCanada
| | - Lirlândia P. Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de FarmáciaUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Katsue Suzuki‐Inoue
- Department of Clinical and Laboratory MedicineUniversity of YamanashiYamanashiJapan
| | - Tahira Zafar
- Frontier Medical CollegeAbbotabadPakistan
- Hemophilia Treatment CenterRawalpindiPakistan
| | - Jiaxi Zhou
- Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| |
Collapse
|
23
|
Ditzig Z, Wilson CM, Salas J, Serve KM. Plasminogen Binding and Activation at the Mesothelial Cell Surface Promotes Invasion through a Collagen Matrix. Int J Mol Sci 2022; 23:ijms23115984. [PMID: 35682663 PMCID: PMC9180734 DOI: 10.3390/ijms23115984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 01/04/2023] Open
Abstract
Plasminogen (Plg) activation to the serine protease plasmin (Pla) plays a key role in regulating wound healing and fibrotic responses, particularly when bound to cell surface receptors. Our previous work suggested that mesothelial cells bind Plg at the cell surface, though no Plg receptors were described for these cells. Since mesothelial cells contribute to injury responses, including cellular differentiation to a mesenchymal-like phenotype and extracellular matrix remodeling, we hypothesized that Plg binding would promote these responses. Here, we confirm that Plg binds to both pleural and peritoneal mesothelial cells via the lysine-binding domain present in Plg, and we demonstrate the presence of three Plg receptors on the mesothelial cell surface: α-Enolase, Annexin A2, and Plg-RKT. We further show that bound-Plg is activated to Pla on the cell surface and that activation is blocked by an inhibitor of urokinase plasminogen activator or by the presence of animal-derived FBS. Lastly, we demonstrate that Plg promotes mesothelial cell invasion through a type I collagen matrix but does not promote cellular differentiation or proliferation. These data demonstrate for the first time that mesothelial cells bind and activate Plg at the cell surface and that active Pla is involved in mesothelial cell invasion without cell differentiation.
Collapse
|
24
|
Li P, Ma C, Li J, You S, Dang L, Wu J, Hao Z, Li J, Zhi Y, Chen L, Sun S. Proteomic characterization of four subtypes of M2 macrophages derived from human THP-1 cells. J Zhejiang Univ Sci B 2022; 23:407-422. [PMID: 35557041 PMCID: PMC9110321 DOI: 10.1631/jzus.b2100930] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/04/2022] [Indexed: 02/06/2023]
Abstract
Macrophages are widely distributed immune cells that contribute to tissue homeostasis. Human THP-1 cells have been widely used in various macrophage-associated studies, especially those involving pro-inflammatory M1 and anti-inflammatory M2 phenotypes. However, the molecular characterization of four M2 subtypes (M2a, M2b, M2c, and M2d) derived from THP-1 has not been fully investigated. In this study, we systematically analyzed the protein expression profiles of human THP-1-derived macrophages (M0, M1, M2a, M2b, M2c, and M2d) using quantitative proteomics approaches. The commonly and specially regulated proteins of the four M2 subtypes and their potential biological functions were further investigated. The results showed that M2a and M2b, and M2c and M2d have very similar protein expression profiles. These data could serve as an important resource for studies of macrophages using THP-1 cells, and provide a reference to distinguish different M2 subtypes in macrophage-associated diseases for subsequent clinical research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Shisheng Sun
- College of Life Science, Northwest University, Xi'an 710069, China.
| |
Collapse
|
25
|
Draxler DF, Hanafi G, Zahra S, McCutcheon F, Ho H, Keragala CB, Liu Z, Daly D, Painter T, Wallace S, Plebanski M, Myles PS, Medcalf RL. Tranexamic acid alters the immunophenotype of phagocytes after lower limb surgery. Thromb J 2022; 20:17. [PMID: 35410340 PMCID: PMC8996554 DOI: 10.1186/s12959-022-00373-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/04/2022] [Indexed: 11/23/2022] Open
Abstract
Background Tranexamic acid (TXA) is an antifibrinolytic agent frequently used in elective surgery to reduce blood loss. We recently found it also acts as a potent immune-modulator in patients undergoing cardiac surgery. Methods Patients undergoing lower limb surgery were enrolled into the “Tranexamic Acid in Lower Limb Arthroplasty” (TALLAS) pilot study. The cellular immune response was characterised longitudinally pre- and post-operatively using full blood examination (FBE) and comprehensive immune cell phenotyping by flowcytometry. Red blood cells and platelets were determined in the FBE and levels of T cell cytokines and the plasmin-antiplasmin complex determined using ELISA. Results TXA administration increased the proportion of circulating CD141+ conventional dendritic cells (cDC) on post-operative day (POD) 3. It also reduced the expression of CD83 and TNFR2 on classical monocytes and levels of circulating IL-10 at the end of surgery (EOS) time point, whilst increasing the expression of CCR4 on natural killer (NK) cells at EOS, and reducing TNFR2 on POD-3 on NK cells. Red blood cells and platelets were decreased to a lower extent at POD-1 in the TXA group, representing reduced blood loss. Conclusion In this investigation we have extended our examination on the immunomodulatory effects of TXA in surgery by also characterising the end of surgery time point and including B cells and neutrophils in our immune analysis, elucidating new immunophenotypic changes in phagocytes as well as NK cells. This study enhances our understanding of TXA-mediated effects on the haemostatic and immune response in surgery, validating changes in important functional immune cell subsets in orthopaedic patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12959-022-00373-3.
Collapse
|
26
|
Miles LA, Krajewski S, Baik N, Parmer RJ, Mueller BM. Plg-RKT Expression in Human Breast Cancer Tissues. Biomolecules 2022; 12:biom12040503. [PMID: 35454092 PMCID: PMC9028288 DOI: 10.3390/biom12040503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
The plasminogen activation system regulates the activity of the serine protease, plasmin. The role of plasminogen receptors in cancer progression is being increasingly appreciated as key players in modulation of the tumor microenvironment. The interaction of plasminogen with cells to promote plasminogen activation requires the presence of proteins exposing C-terminal lysines on the cell surface. Plg-RKT is a structurally unique plasminogen receptor because it is an integral membrane protein that is synthesized with and binds plasminogen via a C-terminal lysine exposed on the cell surface. Here, we have investigated the expression of Plg-RKT in human breast tumors and human breast cancer cell lines. Breast cancer progression tissue microarrays were probed with anti-Plg-RKT mAB and we found that Plg-RKT is widely expressed in human breast tumors, that its expression is increased in tumors that have spread to draining lymph nodes and distant organs, and that Plg-RKT expression is most pronounced in hormone receptor (HR)-positive tumors. Plg-RKT was detected by Western blotting in human breast cancer cell lines. By flow cytometry, Plg-RKT cell surface expression was highest on the most aggressive tumor cell line. Future studies are warranted to address the functions of Plg-RKT in breast cancer.
Collapse
Affiliation(s)
- Lindsey A. Miles
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (L.A.M.); (N.B.)
| | | | - Nagyung Baik
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA; (L.A.M.); (N.B.)
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare System, University of California San Diego, San Diego, CA 92161, USA;
| | - Barbara M. Mueller
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
- Correspondence:
| |
Collapse
|
27
|
Samad F, Bai H, Baik N, Haider P, Zhang Y, Rega-Kaun G, Kaun C, Prager M, Wojta J, Bui Q, Chakrabarty S, Wang J, Parmer RJ, Miles LA. The plasminogen receptor Plg-R KT regulates adipose function and metabolic homeostasis. J Thromb Haemost 2022; 20:742-754. [PMID: 34897983 PMCID: PMC8885904 DOI: 10.1111/jth.15622] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 11/30/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Plg-RKT , a unique transmembrane plasminogen receptor, enhances the activation of plasminogen to plasmin, and localizes the proteolytic activity of plasmin on the cell surface. OBJECTIVES We investigated the role of Plg-RKT in adipose function, metabolic homeostasis, and obesity. METHODS We used adipose tissue (AT) sections from bariatric surgery patients and from high fat diet (HFD)-induced obese mice together with immunofluorescence and real-time polymerase chain reaction to study adipose expression of Plg-RKT . Mice genetically deficient in Plg-RKT and littermate controls fed a HFD or control low fat diet (LFD) were used to determine the role of Plg-RKT in insulin resistance, glucose tolerance, type 2 diabetes, and associated mechanisms including adipose inflammation, fibrosis, and ectopic lipid storage. The role of Plg-RKT in adipogenesis was determined using 3T3-L1 preadipocytes and primary cultures established from Plg-RKT -deficient and littermate control mice. RESULTS Plg-RKT was highly expressed in both human and mouse AT, and its levels dramatically increased during adipogenesis. Plg-RKT -deficient mice, when fed a HFD, gained more weight, developed more hepatic steatosis, and were more insulin resistant/glucose intolerant than HFD-fed wild-type littermates. Mechanistically, these metabolic defects were linked with increased AT inflammation, AT macrophage and T-cell accumulation, adipose and hepatic fibrosis, and decreased insulin signaling in the AT and liver. Moreover, Plg-RKT regulated the expression of PPARγ and other adipogenic molecules, suggesting a novel role for Plg-RKT in the adipogenic program. CONCLUSIONS Plg-RKT coordinately regulates multiple aspects of adipose function that are important to maintain efficient metabolic homeostasis.
Collapse
Affiliation(s)
- Fahumiya Samad
- Department of Cell Biology, San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Hongdong Bai
- Department of Medicine, Veterans Administration San Diego Healthcare System, San Diego
| | - Nagyung Baik
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Patrick Haider
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Yuqing Zhang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Gersina Rega-Kaun
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- 5th Department of Internal Medicine for Diabetes and Rheumatology, Wilhelminen Hospital, Vienna, Austria
| | - Christoph Kaun
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Manfred Prager
- Department of Surgery, Hospital Oberwart, Oberwart, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna
| | - Quyen Bui
- Department of Cell Biology, San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Sagarika Chakrabarty
- Department of Cell Biology, San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Jing Wang
- Department of Cell Biology, San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare System, San Diego
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Lindsey A. Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
28
|
Wysmołek ME, Długosz E, Wiśniewski M. The Immunological Role of Vascular and Lymphatic Endothelial Cells in Filarial Infections. Animals (Basel) 2022; 12:ani12040426. [PMID: 35203133 PMCID: PMC8868237 DOI: 10.3390/ani12040426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/25/2022] [Accepted: 02/07/2022] [Indexed: 11/26/2022] Open
Abstract
Simple Summary The endothelium is a monolayer of cells forming a thin membrane that lines the inside of blood vessels. These cells release molecules that regulate vascular relaxation, contraction, and can control blood clotting and the immune response. During infections with filarial nematodes, common parasites of humans and animals, the endothelium is believed to play a key role in the communication between the host and the parasite, since the embryonic stage of filaroids is distributed in the bloodstream. Therefore, this review aims to gather research from different scientists in order to better understand the host immune response in infections with filarial nematodes. Abstract The embryonic stage of filarial nematodes, or microfilariae (Mf), shows daily and seasonal periodicity that requires their migration through blood vessels into the lungs, where they are sequestered when not circulating in the peripheral blood. Therefore, Mf and the host endothelium are likely in a permanent state of hide and seek. Interestingly, filarial nematodes co-cultured in media with a murine endothelial cell line survive eight times longer than those cultured in media alone. This suggests that the endothelium is an important element of the immune response in filarial nematodes, perversely promoting their survival in the host. In this review, we will focus on potential pathways involved in the relationship between filarial nematodes and the host endothelium, including the role of endothelial ICAM/VCAM/PECAM adhesion molecules, surface markers involved in the passage of Mf through host tissue, anti-thrombolic effects caused by the presence of filarial nematodes (including plasmins), endothelial cell proliferation (VEGF), and other aspects of the immune activation of the endothelium. The aim of this review is to merge the knowledge about the cross-talk between Mf of different filarial nematode species and endothelial cells (EC), thus allowing a better understanding of the mechanism of these parasitic infections.
Collapse
|
29
|
Coagulome and the tumor microenvironment: an actionable interplay. Trends Cancer 2022; 8:369-383. [PMID: 35027336 DOI: 10.1016/j.trecan.2021.12.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/19/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022]
Abstract
Human tumors often trigger a hypercoagulable state that promotes hemostatic complications, including venous thromboembolism. The recent application of systems biology to the study of the coagulome highlighted its link to shaping the tumor microenvironment (TME), both within and outside of the vascular space. Addressing this link provides the opportunity to revisit the significance of biomarkers of hemostasis and assess the communication between vasculature and tumor parenchyma, an important topic considering the advent of immune checkpoint inhibitors and vascular normalization strategies. Understanding how the coagulome and TME influence each other offers exciting new prospects for predicting hemostatic complications and boosting the effectiveness of cancer treatment.
Collapse
|
30
|
Gibson BHY, Wollenman CC, Moore-Lotridge SN, Keller PR, Summitt JB, Revenko AR, Flick MJ, Blackwell TS, Schoenecker JG. Plasmin drives burn-induced systemic inflammatory response syndrome. JCI Insight 2021; 6:154439. [PMID: 34877937 PMCID: PMC8675186 DOI: 10.1172/jci.insight.154439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
Severe injuries, such as burns, provoke a systemic inflammatory response syndrome (SIRS) that imposes pathology on all organs. Simultaneously, severe injury also elicits activation of the fibrinolytic protease plasmin. While the principal adverse outcome of plasmin activation in severe injury is compromised hemostasis, plasmin also possesses proinflammatory properties. We hypothesized that, following a severe injury, early activation of plasmin drives SIRS. Plasmin activation was measured and related to injury severity, SIRS, coagulopathy, and outcomes prospectively in burn patients who are not at risk of hemorrhage. Patients exhibited early, significant activation of plasmin that correlated with burn severity, cytokines, coagulopathy, and death. Burn with a concomitant, remote muscle injury was employed in mice to determine the role of plasmin in the cytokine storm and inflammatory cascades in injured tissue distant from the burn injury. Genetic and pharmacologic inhibition of plasmin reduced the burn-induced cytokine storm and inflammatory signaling in injured tissue. These findings demonstrate (a) that severe injury-induced plasmin activation is a key pathologic component of the SIRS-driven cytokine storm and SIRS-activated inflammatory cascades in tissues distant from the inciting injury and (b) that targeted inhibition of plasmin activation may be effective for limiting both hemorrhage and tissue-damaging inflammation following injury.
Collapse
Affiliation(s)
| | - Colby C Wollenman
- School of Medicine.,Department of Orthopaedic Surgery, Vanderbilt University Medical Center
| | - Stephanie N Moore-Lotridge
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center.,Vanderbilt Center for Bone Biology
| | | | - J Blair Summitt
- Department of Plastic Surgery, Vanderbilt University Medical Center; and.,Vanderbilt University Medical Center Burn Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Alexey R Revenko
- IONIS Pharmaceuticals Pulmonary and Oncology Drug Discovery, Carlsbad, California, USA
| | - Matthew J Flick
- Department of Pathology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA.,University of North Carolina Blood Research Center, Chapel Hill, North Carolina, USA
| | - Timothy S Blackwell
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA.,Division of Pulmonary and Critical Care
| | - Jonathan G Schoenecker
- Department of Pharmacology.,Department of Orthopaedic Surgery, Vanderbilt University Medical Center.,Vanderbilt Center for Bone Biology.,Department of Pathology, Microbiology, and Immunology; and.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
31
|
Zaidan I, Tavares LP, Sugimoto MA, Lima KM, Negreiros-Lima GL, Teixeira LC, Miranda TC, Valiate BV, Cramer A, Vago JP, Campolina-Silva GH, Souza JA, Grossi LC, Pinho V, Campagnole-Santos MJ, Santos RAS, Teixeira MM, Galvão I, Sousa LP. Angiotensin-(1-7)/MasR axis promotes migration of monocytes/macrophages with a regulatory phenotype to perform phagocytosis and efferocytosis. JCI Insight 2021; 7:147819. [PMID: 34874920 PMCID: PMC8765051 DOI: 10.1172/jci.insight.147819] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Nonphlogistic migration of macrophages contributes to the clearance of pathogens and apoptotic cells, a critical step for the resolution of inflammation and return to homeostasis. Angiotensin-(1-7) [Ang-(1-7)] is a heptapeptide of the renin-angiotensin system that acts through Mas receptor (MasR). Ang-(1-7) has recently emerged as a novel proresolving mediator, yet Ang-(1-7) resolution mechanisms are not fully determined. Herein, Ang-(1-7) stimulated migration of human and murine monocytes/macrophages in a MasR-, CCR2-, and MEK/ERK1/2–dependent manner. Pleural injection of Ang-(1-7) promoted nonphlogistic mononuclear cell influx alongside increased levels of CCL2, IL-10, and macrophage polarization toward a regulatory phenotype. Ang-(1-7) induction of CCL2 and mononuclear cell migration was also dependent on MasR and MEK/ERK. Of note, MasR was upregulated during the resolution phase of inflammation, and its pharmacological inhibition or genetic deficiency impaired mononuclear cell recruitment during self-resolving models of LPS pleurisy and E. coli peritonitis. Inhibition/absence of MasR was associated with reduced CCL2 levels, impaired phagocytosis of bacteria, efferocytosis, and delayed resolution of inflammation. In summary, we have uncovered a potentially novel proresolving feature of Ang-(1-7), namely the recruitment of mononuclear cells favoring efferocytosis, phagocytosis, and resolution of inflammation. Mechanistically, cell migration was dependent on MasR, CCR2, and the MEK/ERK pathway.
Collapse
Affiliation(s)
- Isabella Zaidan
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana P Tavares
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michelle A Sugimoto
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kátia M Lima
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Graziele L Negreiros-Lima
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lívia Cr Teixeira
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thais C Miranda
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno Vs Valiate
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Allysson Cramer
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Priscila Vago
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Jéssica Am Souza
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laís C Grossi
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Robson A S Santos
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela Galvão
- Departamento Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Department of Clinical and Toxicological Analysis from the School of Pharma, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
32
|
Guzman RM, Howard ZP, Liu Z, Oliveira RD, Massa AT, Omsland A, White SN, Goodman AG. Natural genetic variation in Drosophila melanogaster reveals genes associated with Coxiella burnetii infection. Genetics 2021; 217:6117219. [PMID: 33789347 PMCID: PMC8045698 DOI: 10.1093/genetics/iyab005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/07/2021] [Indexed: 12/16/2022] Open
Abstract
The gram-negative bacterium Coxiella burnetii is the causative agent of Query (Q) fever in humans and coxiellosis in livestock. Host genetics are associated with C. burnetii pathogenesis both in humans and animals; however, it remains unknown if specific genes are associated with severity of infection. We employed the Drosophila Genetics Reference Panel to perform a genome-wide association study to identify host genetic variants that affect host survival to C. burnetii infection. The genome-wide association study identified 64 unique variants (P < 10−5) associated with 25 candidate genes. We examined the role each candidate gene contributes to host survival during C. burnetii infection using flies carrying a null mutation or RNAi knockdown of each candidate. We validated 15 of the 25 candidate genes using at least one method. This is the first report establishing involvement of many of these genes or their homologs with C. burnetii susceptibility in any system. Among the validated genes, FER and tara play roles in the JAK/STAT, JNK, and decapentaplegic/TGF-β signaling pathways which are components of known innate immune responses to C. burnetii infection. CG42673 and DIP-ε play roles in bacterial infection and synaptic signaling but have no previous association with C. burnetii pathogenesis. Furthermore, since the mammalian ortholog of CG13404 (PLGRKT) is an important regulator of macrophage function, CG13404 could play a role in host susceptibility to C. burnetii through hemocyte regulation. These insights provide a foundation for further investigation regarding the genetics of C. burnetii susceptibility across a wide variety of hosts.
Collapse
Affiliation(s)
- Rosa M Guzman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Zachary P Howard
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Ziying Liu
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Ryan D Oliveira
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Alisha T Massa
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Anders Omsland
- Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Stephen N White
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.,USDA-ARS Animal Disease Research, Pullman, WA 99164, USA.,Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - Alan G Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.,Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
33
|
Integrated Metabolomics and Proteomics Analyses in the Local Milieu of Islet Allografts in Rejection versus Tolerance. Int J Mol Sci 2021; 22:ijms22168754. [PMID: 34445459 PMCID: PMC8395897 DOI: 10.3390/ijms22168754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 11/24/2022] Open
Abstract
An understanding of the immune mechanisms that lead to rejection versus tolerance of allogeneic pancreatic islet grafts is of paramount importance, as it facilitates the development of innovative methods to improve the transplant outcome. Here, we used our established intraocular islet transplant model to gain novel insight into changes in the local metabolome and proteome within the islet allograft’s immediate microenvironment in association with immune-mediated rejection or tolerance. We performed integrated metabolomics and proteomics analyses in aqueous humor samples representative of the graft’s microenvironment under each transplant outcome. The results showed that several free amino acids, small primary amines, and soluble proteins related to the Warburg effect were upregulated or downregulated in association with either outcome. In general, the observed shifts in the local metabolite and protein profiles in association with rejection were consistent with established pro-inflammatory metabolic pathways and those observed in association with tolerance were immune regulatory. Taken together, the current findings further support the potential of metabolic reprogramming of immune cells towards immune regulation through targeted pharmacological and dietary interventions against specific metabolic pathways that promote the Warburg effect to prevent the rejection of transplanted islets and promote their immune tolerance.
Collapse
|
34
|
Vago JP, Amaral FA, van de Loo FAJ. Resolving inflammation by TAM receptor activation. Pharmacol Ther 2021; 227:107893. [PMID: 33992683 DOI: 10.1016/j.pharmthera.2021.107893] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
The control of inflammation is strictly regulated to ensure the adequate intensity and duration of an inflammatory response, enabling the removal of the trigger factors and the restoration of the integrity of the tissues and their functions. This process is coordinated by anti-inflammatory and pro-resolving mediators that regulate the cellular and molecular events necessary to restore homeostasis, and defects in this control are associated with the development of chronic and autoimmune diseases. The TAM family of receptor tyrosine kinases-Tyro3, Axl, and MerTK-plays an essential role in efferocytosis, a key process for the resolution of inflammation. However, new studies have demonstrated that TAM receptor activation not only reduces the synthesis of pro-inflammatory mediators by different cell types in response to some stimuli but also stimulates the production of anti-inflammatory and pro-resolving molecules that control the inflammation. This review provides a comprehensive view of TAM receptor family members as important players in controlling inflammatory responses through anti-inflammatory and pro-resolving actions.
Collapse
Affiliation(s)
- Juliana P Vago
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, the Netherlands
| | - Flávio A Amaral
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, the Netherlands; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fons A J van de Loo
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, the Netherlands.
| |
Collapse
|
35
|
Felix FB, Vago JP, Fernandes DDO, Martins DG, Moreira IZ, Gonçalves WA, Costa WC, Araújo JMD, Queiroz-Junior CM, Campolina-Silva GH, Soriani FM, Sousa LP, Grespan R, Teixeira MM, Pinho V. Biochanin A Regulates Key Steps of Inflammation Resolution in a Model of Antigen-Induced Arthritis via GPR30/PKA-Dependent Mechanism. Front Pharmacol 2021; 12:662308. [PMID: 33995086 PMCID: PMC8114065 DOI: 10.3389/fphar.2021.662308] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Biochanin A (BCA) is a natural organic compound of the class of phytochemicals known as flavonoids and isoflavone subclass predominantly found in red clover (Trifolium pratense). It has anti-inflammatory activity and some pro-resolving actions, such as neutrophil apoptosis. However, the effect of BCA in the resolution of inflammation is still poorly understood. In this study, we investigated the effects of BCA on the neutrophilic inflammatory response and its resolution in a model of antigen-induced arthritis. Male wild-type BALB/c mice were treated with BCA at the peak of the inflammatory process (12 h). BCA decreased the accumulation of migrated neutrophils, and this effect was associated with reduction of myeloperoxidase activity, IL-1β and CXCL1 levels, and the histological score in periarticular tissues. Joint dysfunction, as seen by mechanical hypernociception, was improved by treatment with BCA. The resolution interval (Ri) was also quantified, defining profiles of acute inflammatory parameters that include the amplitude and duration of the inflammatory response monitored by the neutrophil infiltration. BCA treatment shortened Ri from ∼23 h observed in vehicle-treated mice to ∼5.5 h, associated with an increase in apoptotic events and efferocytosis, both key steps for the resolution of inflammation. These effects of BCA were prevented by H89, an inhibitor of protein kinase A (PKA) and G15, a selective G protein–coupled receptor 30 (GPR30) antagonist. In line with the in vivo data, BCA also increased the efferocytic ability of murine bone marrow–derived macrophages. Collectively, these data indicate for the first time that BCA resolves neutrophilic inflammation acting in key steps of the resolution of inflammation, requiring activation of GPR30 and via stimulation of cAMP-dependent signaling.
Collapse
Affiliation(s)
- Franciel Batista Felix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Priscila Vago
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Débora de Oliveira Fernandes
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Débora Gonzaga Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Zaidan Moreira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - William Antonio Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Walyson Coelho Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Frederico Marianetti Soriani
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia Pires Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Renata Grespan
- Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristovão, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
36
|
Medcalf RL, Keragala CB. Fibrinolysis: A Primordial System Linked to the Immune Response. Int J Mol Sci 2021; 22:3406. [PMID: 33810275 PMCID: PMC8037105 DOI: 10.3390/ijms22073406] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 01/07/2023] Open
Abstract
The fibrinolytic system provides an essential means to remove fibrin deposits and blood clots. The actual protease responsible for this is plasmin, formed from its precursor, plasminogen. Fibrin is heralded as it most renowned substrate but for many years plasmin has been known to cleave many other substrates, and to also activate other proteolytic systems. Recent clinical studies have shown that the promotion of plasmin can lead to an immunosuppressed phenotype, in part via its ability to modulate cytokine expression. Almost all immune cells harbor at least one of a dozen plasminogen receptors that allows plasmin formation on the cell surface that in turn modulates immune cell behavior. Similarly, a multitude of pathogens can also express their own plasminogen activators, or contain surface proteins that provide binding sites host plasminogen. Plasmin formed under these circumstances also empowers these pathogens to modulate host immune defense mechanisms. Phylogenetic studies have revealed that the plasminogen activating system predates the appearance of fibrin, indicating that plasmin did not evolve as a fibrinolytic protease but perhaps has its roots as an immune modifying protease. While its fibrin removing capacity became apparent in lower vertebrates these primitive under-appreciated immune modifying functions still remain and are now becoming more recognised.
Collapse
Affiliation(s)
- Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis Laboratory, Australian Centre for Blood Diseases, Central Clinical School Melbourne, Monash University, Melbourne, VIC 3004, Australia;
| | | |
Collapse
|
37
|
Yaron JR, Zhang L, Guo Q, Haydel SE, Lucas AR. Fibrinolytic Serine Proteases, Therapeutic Serpins and Inflammation: Fire Dancers and Firestorms. Front Cardiovasc Med 2021; 8:648947. [PMID: 33869309 PMCID: PMC8044766 DOI: 10.3389/fcvm.2021.648947] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The making and breaking of clots orchestrated by the thrombotic and thrombolytic serine protease cascades are critical determinants of morbidity and mortality during infection and with vascular or tissue injury. Both the clot forming (thrombotic) and the clot dissolving (thrombolytic or fibrinolytic) cascades are composed of a highly sensitive and complex relationship of sequentially activated serine proteases and their regulatory inhibitors in the circulating blood. The proteases and inhibitors interact continuously throughout all branches of the cardiovascular system in the human body, representing one of the most abundant groups of proteins in the blood. There is an intricate interaction of the coagulation cascades with endothelial cell surface receptors lining the vascular tree, circulating immune cells, platelets and connective tissue encasing the arterial layers. Beyond their role in control of bleeding and clotting, the thrombotic and thrombolytic cascades initiate immune cell responses, representing a front line, "off-the-shelf" system for inducing inflammatory responses. These hemostatic pathways are one of the first response systems after injury with the fibrinolytic cascade being one of the earliest to evolve in primordial immune responses. An equally important contributor and parallel ancient component of these thrombotic and thrombolytic serine protease cascades are the serine protease inhibitors, termed serpins. Serpins are metastable suicide inhibitors with ubiquitous roles in coagulation and fibrinolysis as well as multiple central regulatory pathways throughout the body. Serpins are now known to also modulate the immune response, either via control of thrombotic and thrombolytic cascades or via direct effects on cellular phenotypes, among many other functions. Here we review the co-evolution of the thrombolytic cascade and the immune response in disease and in treatment. We will focus on the relevance of these recent advances in the context of the ongoing COVID-19 pandemic. SARS-CoV-2 is a "respiratory" coronavirus that causes extensive cardiovascular pathogenesis, with microthrombi throughout the vascular tree, resulting in severe and potentially fatal coagulopathies.
Collapse
Affiliation(s)
- Jordan R. Yaron
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School for Engineering of Matter, Transport and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, United States
| | - Liqiang Zhang
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Qiuyun Guo
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Shelley E. Haydel
- Center for Bioelectronics and Biosensors, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
38
|
Miles LA, Ny L, Wilczynska M, Shen Y, Ny T, Parmer RJ. Plasminogen Receptors and Fibrinolysis. Int J Mol Sci 2021; 22:ijms22041712. [PMID: 33567773 PMCID: PMC7914795 DOI: 10.3390/ijms22041712] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
The ability of cells to promote plasminogen activation on their surfaces is now well recognized, and several distinct cell surface proteins have been demonstrated to function as plasminogen receptors. Here, we review studies demonstrating that plasminogen bound to cells, in addition to plasminogen directly bound to fibrin, plays a major role in regulating fibrin surveillance. We focus on the ability of specific plasminogen receptors on eukaryotic cells to promote fibrinolysis in the in vivo setting by reviewing data obtained predominantly in murine models. Roles for distinct plasminogen receptors in fibrin surveillance in intravascular fibrinolysis, immune cell recruitment in the inflammatory response, wound healing, and lactational development are discussed.
Collapse
Affiliation(s)
- Lindsey A. Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Correspondence: ; Tel.: +1-858-784-7105; Fax: 858-784-7374
| | - Lina Ny
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden; (L.N.); (M.W.); (Y.S.); (T.N.)
| | - Malgorzata Wilczynska
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden; (L.N.); (M.W.); (Y.S.); (T.N.)
| | - Yue Shen
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden; (L.N.); (M.W.); (Y.S.); (T.N.)
| | - Tor Ny
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187 Umeå, Sweden; (L.N.); (M.W.); (Y.S.); (T.N.)
| | - Robert J. Parmer
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA and Veterans Administration San Diego Healthcare System, San Diego, CA 92161, USA;
| |
Collapse
|
39
|
Whyte CS, Morrow GB, Baik N, Booth NA, Jalal MM, Parmer RJ, Miles LA, Mutch NJ. Exposure of plasminogen and a novel plasminogen receptor, Plg-RKT, on activated human and murine platelets. Blood 2021; 137:248-257. [PMID: 32842150 PMCID: PMC7820873 DOI: 10.1182/blood.2020007263] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/15/2020] [Indexed: 11/20/2022] Open
Abstract
Plasminogen activation rates are enhanced by cell surface binding. We previously demonstrated that exogenous plasminogen binds to phosphatidylserine-exposing and spread platelets. Platelets contain plasminogen in their α-granules, but secretion of plasminogen from platelets has not been studied. Recently, a novel transmembrane lysine-dependent plasminogen receptor, Plg-RKT, has been described on macrophages. Here, we analyzed the pool of plasminogen in platelets and examined whether platelets express Plg-RKT. Plasminogen content of the supernatant of resting and collagen/thrombin-stimulated platelets was similar. Pretreatment with the lysine analog, ε-aminocaproic acid, significantly increased platelet-derived plasminogen (0.33 vs 0.08 nmol/108 platelets) in the stimulated supernatant, indicating a lysine-dependent mechanism of membrane retention. Lysine-dependent, platelet-derived plasminogen retention on thrombin and convulxin activated human platelets was confirmed by flow cytometry. Platelets initiated fibrinolytic activity in fluorescently labeled plasminogen-deficient clots and in turbidimetric clot lysis assays. A 17-kDa band, consistent with Plg-RKT, was detected in the platelet membrane fraction by western blotting. Confocal microscopy of stimulated platelets revealed Plg-RKT colocalized with platelet-derived plasminogen on the activated platelet membrane. Plasminogen exposure was significantly attenuated in thrombin- and convulxin-stimulated platelets from Plg-RKT-/- mice compared with Plg-RKT+/+ littermates. Membrane exposure of Plg-RKT was not dependent on plasminogen, as similar levels of the receptor were detected in plasminogen-/- platelets. These data highlight Plg-RKT as a novel plasminogen receptor in human and murine platelets. We show for the first time that platelet-derived plasminogen is retained on the activated platelet membrane and drives local fibrinolysis by enhancing cell surface-mediated plasminogen activation.
Collapse
Affiliation(s)
- Claire S Whyte
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gael B Morrow
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Nagyung Baik
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Nuala A Booth
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Mohammed M Jalal
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Robert J Parmer
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
- Department of Medicine, University of California San Diego, San Diego, CA; and
| | - Lindsey A Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Nicola J Mutch
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
40
|
Ny L, Parmer RJ, Shen Y, Holmberg S, Baik N, Bäckman A, Broden J, Wilczynska M, Ny T, Miles LA. The plasminogen receptor, Plg-R KT, plays a role in inflammation and fibrinolysis during cutaneous wound healing in mice. Cell Death Dis 2020; 11:1054. [PMID: 33311441 PMCID: PMC7733490 DOI: 10.1038/s41419-020-03230-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Wound healing is a complex physiologic process that proceeds in overlapping, sequential steps. Plasminogen promotes fibrinolysis and potentiates the inflammatory response during wound healing. We have tested the hypothesis that the novel plasminogen receptor, Plg-RKT, regulates key steps in wound healing. Standardized burn wounds were induced in mice and time dependence of wound closure was quantified. Healing in Plg-RKT−/− mice was significantly delayed during the proliferation phase. Expression of inflammatory cytokines was dysregulated in Plg-RKT−/− wound tissue. Consistent with dysregulated cytokine expression, a significant delay in wound healing during the proliferation phase was observed in mice in which Plg-RKT was specifically deleted in myeloid cells. Following wound closure, the epidermal thickness was less in Plg-RKT−/− wound tissue. Paradoxically, deletion of Plg-RKT, specifically in keratinocytes, significantly accelerated the rate of healing during the proliferation phase. Mechanistically, only two genes were upregulated in Plg-RKT−/− compared with Plg-RKT+/+ wound tissue, filaggrin, and caspase 14. Both filaggrin and caspase 14 promote epidermal differentiation and decrease proliferation, consistent with more rapid wound closure and decreased epidermal thickness during the remodeling phase. Fibrin clearance was significantly impaired in Plg-RKT−/− wound tissue. Genetic reduction of fibrinogen levels to 50% completely abrogated the effect of Plg-RKT deletion on the healing of burn wounds. Remarkably, the effects of Plg-RKT deletion on cytokine expression were modulated by reducing fibrinogen levels. In summary, Plg-RKT is a new regulator participating in different phases of cutaneous burn wound healing, which coordinately plays a role in the interrelated responses of inflammation, keratinocyte migration, and fibrinolysis.
Collapse
Affiliation(s)
- Lina Ny
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | - Robert J Parmer
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.,Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Yue Shen
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | | | - Nagyung Baik
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | - Malgorzata Wilczynska
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | - Tor Ny
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
| | - Lindsey A Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
41
|
Sharma S, Shinde SS, Teekas L, Vijay N. Evidence for the loss of plasminogen receptor KT gene in chicken. Immunogenetics 2020; 72:507-515. [PMID: 33247773 DOI: 10.1007/s00251-020-01186-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/03/2020] [Indexed: 12/31/2022]
Abstract
The loss of conserved genes has the potential to alter phenotypes drastically. Screening of vertebrate genomes for lineage-specific gene loss events has identified numerous natural knockouts associated with specific phenotypes. We provide evidence for the loss of a multi-exonic plasminogen receptor KT (PLGRKT) protein-encoding gene located on the Z chromosome in chicken. Exons 1 and 2 are entirely missing; remnants of exon 3 and a mostly intact exon 4 are identified in an assembly gap-free region in chicken with conserved synteny across species and verified using transcriptome and genome sequencing. PLGRKT gene disrupting changes are present in representative species from all five galliform families. In contrast to this, the presence of an intact transcriptionally active PLGRKT gene in species such as mallard, swan goose, and Anolis lizard suggests that gene loss occurred in the galliform lineage sometime between 68 and 80 Mya. The presence of galliform specific chicken repeat 1 (CR1) insertion at the erstwhile exon 2 of PLGRKT gene suggests repeat insertion-mediated loss. However, at least nine other independent PLGRKT coding frame disrupting changes in other bird species are supported by genome sequencing and indicate a role for relaxed purifying selection before CR1 insertion. The recurrent loss of a conserved gene with a role in the regulation of macrophage migration, efferocytosis, and blood coagulation is intriguing. Hence, we propose potential candidate genes that might be compensating the function of PLGRKT based on the presence of a C-terminal lysine residue, transmembrane domains, and gene expression patterns.
Collapse
Affiliation(s)
- Sandhya Sharma
- Computational Evolutionary Genomics Lab, Department of Biological Sciences, IISER Bhopal, Bhauri, Madhya Pradesh, India
| | - Sagar Sharad Shinde
- Computational Evolutionary Genomics Lab, Department of Biological Sciences, IISER Bhopal, Bhauri, Madhya Pradesh, India
| | - Lokdeep Teekas
- Computational Evolutionary Genomics Lab, Department of Biological Sciences, IISER Bhopal, Bhauri, Madhya Pradesh, India
| | - Nagarjun Vijay
- Computational Evolutionary Genomics Lab, Department of Biological Sciences, IISER Bhopal, Bhauri, Madhya Pradesh, India.
| |
Collapse
|
42
|
Repurposing of Plasminogen: An Orphan Medicinal Product Suitable for SARS-CoV-2 Inhalable Therapeutics. Pharmaceuticals (Basel) 2020; 13:ph13120425. [PMID: 33260813 PMCID: PMC7761183 DOI: 10.3390/ph13120425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
The SARS-CoV-2 infection is associated with pulmonary coagulopathy, which determines the deposition of fibrin in the air spaces and lung parenchyma. The resulting lung lesions compromise patient pulmonary function and increase mortality, or end in permanent lung damage for those who have recovered from the COVID-19 disease. Therefore, local pulmonary fibrinolysis can be efficacious in degrading pre-existing fibrin clots and reducing the conversion of lung lesions into lasting scars. Plasminogen is considered a key player in fibrinolysis processes, and in view of a bench-to-bedside translation, we focused on the aerosolization of an orphan medicinal product (OMP) for ligneous conjunctivitis: human plasminogen (PLG-OMP) eye drops. As such, the sterile and preservative-free solution guarantees the pharmaceutical quality of GMP production and meets the Ph. Eur. requirements of liquid preparations for nebulization. PLG-OMP aerosolization was evaluated both from technological and stability viewpoints, after being submitted to either jet or ultrasonic nebulization. Jet nebulization resulted in a more efficient delivery of an aerosol suitable for pulmonary deposition. The biochemical investigation highlighted substantial protein integrity maintenance with the percentage of native plasminogen band > 90%, in accordance with the quality specifications of PLG-OMP. In a coherent way, the specific activity of plasminogen is maintained within the range 4.8–5.6 IU/mg (PLG-OMP pre-nebulization: 5.0 IU/mg). This is the first study that focuses on the technological and biochemical aspects of aerosolized plasminogen, which could affect both treatment efficacy and clinical dosage delivery. Increasing evidence for the need of local fibrinolytic therapy could merge with the availability of PLG-OMP as an easy handling solution, readily aerosolizable for a fast translation into an extended clinical efficacy assessment in COVID-19 patients.
Collapse
|
43
|
Baker SK, Strickland S. A critical role for plasminogen in inflammation. J Exp Med 2020; 217:133866. [PMID: 32159743 PMCID: PMC7144526 DOI: 10.1084/jem.20191865] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/10/2019] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Plasminogen and its active form, plasmin, have diverse functions related to the inflammatory response in mammals. Due to these roles in inflammation, plasminogen has been implicated in the progression of a wide range of diseases with an inflammatory component. In this review, we discuss the functions of plasminogen in inflammatory regulation and how this system plays a role in the pathogenesis of diseases spanning organ systems throughout the body.
Collapse
Affiliation(s)
- Sarah K Baker
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY
| |
Collapse
|
44
|
Heissig B, Salama Y, Takahashi S, Osada T, Hattori K. The multifaceted role of plasminogen in inflammation. Cell Signal 2020; 75:109761. [PMID: 32861744 PMCID: PMC7452830 DOI: 10.1016/j.cellsig.2020.109761] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 01/01/2023]
Abstract
A fine-tuned activation and deactivation of proteases and their inhibitors are involved in the execution of the inflammatory response. The zymogen/proenzyme plasminogen is converted to the serine protease plasmin, a key fibrinolytic factor by plasminogen activators including tissue-type plasminogen activator (tPA). Plasmin is part of an intricate protease network controlling proteins of initial hemostasis/coagulation, fibrinolytic and complement system. Activation of these protease cascades is required to mount a proper inflammatory response. Although best known for its ability to dissolve clots and cleave fibrin, recent studies point to the importance of fibrin-independent functions of plasmin during acute inflammation and inflammation resolution. In this review, we provide an up-to-date overview of the current knowledge of the enzymatic and cytokine-like effects of tPA and describe the role of tPA and plasminogen receptors in the regulation of the inflammatory response with emphasis on the cytokine storm syndrome such as observed during coronavirus disease 2019 or macrophage activation syndrome. We discuss tPA as a modulator of Toll like receptor signaling, plasmin as an activator of NFkB signaling, and summarize recent studies on the role of plasminogen receptors as controllers of the macrophage conversion into the M2 type and as mediators of efferocytosis during inflammation resolution.
Collapse
Affiliation(s)
- Beate Heissig
- Department of Immunological Diagnosis, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan.
| | - Yousef Salama
- An-Najah Center for Cancer and Stem Cell Research, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Satoshi Takahashi
- Department of Hematology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | - Taro Osada
- Department of Gastroenterology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi, 279-0021 Chiba, Japan.
| | - Koichi Hattori
- Center for Genomic & Regenerative Medicine, Juntendo University, School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan.
| |
Collapse
|
45
|
Miles LA, Vago JP, Sousa LP, Parmer RJ. Functions of the plasminogen receptor Plg-R KT. J Thromb Haemost 2020; 18:2468-2481. [PMID: 32662180 PMCID: PMC7722214 DOI: 10.1111/jth.15014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
Plg-RKT is a structurally unique transmembrane plasminogen receptor with both N- and C-terminal domains exposed on the extracellular face of the cell. Its C-terminal lysine functions to tether plasminogen to cell surfaces. Overexpression of Plg-RKT increases cell surface plasminogen binding capacity while genetic deletion of Plg-RKT decreases plasminogen binding. Plasminogen binding to Plg-RKT results in promotion of plasminogen activation to the broad spectrum serine protease plasmin. This function is promoted by the physical association of Plg-RKT with the urokinase receptor (uPAR). Plg-RKT is broadly expressed in cells and tissues throughout the organism and its sequence is remarkably conserved phylogenetically. Plg-RKT also is required for lactation and, thus, is necessary for survival of the species. This review provides an overview of established and emerging functions of Plg-RKT and highlights major roles for Plg-RKT in both the initiation and resolution of inflammation. While the roles for Plg-RKT in the inflammatory response are predominantly plasmin(ogen)-dependent, its role in lactation requires both plasminogen-dependent and plasminogen-independent mechanisms. Furthermore, the functions of Plg-RKT are dependent on sex. In view of the broad tissue distribution of Plg-RKT , its role in a broad array of physiological and pathological processes should provide a fruitful area for future investigation.
Collapse
Affiliation(s)
- Lindsey A. Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Juliana P. Vago
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P. Sousa
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Robert J. Parmer
- Department of Medicine, University of California San Diego, La Jolla, CA
- Veterans Administration San Diego Healthcare System, San Diego, CA
| |
Collapse
|
46
|
Medcalf RL, Keragala CB, Myles PS. Fibrinolysis and COVID-19: A plasmin paradox. J Thromb Haemost 2020; 18:2118-2122. [PMID: 32543119 PMCID: PMC7323332 DOI: 10.1111/jth.14960] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 12/24/2022]
Abstract
The COVID-19 pandemic has provided many challenges in the field of thrombosis and hemostasis. Among these is a novel form of coagulopathy that includes exceptionally high levels of D-dimer. D-dimer is a marker of poor prognosis, but does this also imply a causal relationship? These spectacularly raised D-dimer levels may actually signify the failing attempt of the fibrinolytic system to remove fibrin and necrotic tissue from the lung parenchyma, being consumed or overwhelmed in the process. Indeed, recent studies suggest that increasing fibrinolytic activity might offer hope for patients with critical disease and severe respiratory failure. However, the fibrinolytic system can also be harnessed by coronavirus to promote infectivity and where antifibrinolytic measures would also seem appropriate. Hence, there is a clinical paradox where plasmin formation can be either deleterious or beneficial in COVID-19, but not at the same time. Hence, it all comes down to timing.
Collapse
Affiliation(s)
- Robert L Medcalf
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Charithani B Keragala
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Paul S Myles
- Department of Anaesthesiology and Perioperative Medicine, Alfred Hospital, Melbourne, Victoria, Australia
- Department of Anaesthesiology and Perioperative Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
47
|
Sousa LP, Pinho V, Teixeira MM. Harnessing inflammation resolving-based therapeutic agents to treat pulmonary viral infections: What can the future offer to COVID-19? Br J Pharmacol 2020; 177:3898-3904. [PMID: 32557557 PMCID: PMC7323156 DOI: 10.1111/bph.15164] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammation is generally accepted as a component of the host defence system and a protective response in the context of infectious diseases. However, altered inflammatory responses can contribute to disease in infected individuals. Many endogenous mediators that drive the resolution of inflammation are now known. Overall, mediators of resolution tend to decrease inflammatory responses and provide normal or greater ability of the host to deal with infection. In the lung, it seems that pro‐resolution molecules, or strategies that promote their increase, tend to suppress inflammation and lung injury and facilitate control of bacterial or viral burden. Here, we argue that the demonstrated anti‐inflammatory, pro‐resolving, anti‐thrombogenic and anti‐microbial effects of such endogenous mediators of resolution may be useful in the treatment of the late stages of the disease in patients with COVID‐19.
Collapse
Affiliation(s)
- Lirlândia P Sousa
- Laboratorio de Imunofamacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Laboratorio de Imunofamacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Laboratorio de Imunofamacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
48
|
Tavares LP, Negreiros-Lima GL, Lima KM, E Silva PMR, Pinho V, Teixeira MM, Sousa LP. Blame the signaling: Role of cAMP for the resolution of inflammation. Pharmacol Res 2020; 159:105030. [PMID: 32562817 DOI: 10.1016/j.phrs.2020.105030] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/06/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
A complex intracellular signaling governs different cellular responses in inflammation. Extracellular stimuli are sensed, amplified, and transduced through a dynamic cellular network of messengers converting the first signal into a proper response: production of specific mediators, cell activation, survival, or death. Several overlapping pathways are coordinated to ensure specific and timely induction of inflammation to neutralize potential harms to the tissue. Ideally, the inflammatory response must be controlled and self-limited. Resolution of inflammation is an active process that culminates with termination of inflammation and restoration of tissue homeostasis. Comparably to the onset of inflammation, resolution responses are triggered by coordinated intracellular signaling pathways that transduce the message to the nucleus. However, the key messengers and pathways involved in signaling transduction for resolution are still poorly understood in comparison to the inflammatory network. cAMP has long been recognized as an inducer of anti-inflammatory responses and cAMP-dependent pathways have been extensively exploited pharmacologically to treat inflammatory diseases. Recently, cAMP has been pointed out as coordinator of key steps of resolution of inflammation. Here, we summarize the evidence for the role of cAMP at inducing important features of resolution of inflammation.
Collapse
Affiliation(s)
- Luciana P Tavares
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA..
| | - Graziele L Negreiros-Lima
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| | - Kátia M Lima
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Post-Graduation Program in Pharmaceutical Sciences, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| | - Patrícia M R E Silva
- Inflammation Laboratory, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.
| | - Vanessa Pinho
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Department of Morphology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil.
| | - Mauro M Teixeira
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil.
| | - Lirlândia P Sousa
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, UFMG, Belo Horizonte, Brazil; Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil; Post-Graduation Program in Pharmaceutical Sciences, Faculdade de Farmácia, UFMG, Belo Horizonte, Brazil.
| |
Collapse
|
49
|
Vago JP, Galvão I, Negreiros-Lima GL, Teixeira LCR, Lima KM, Sugimoto MA, Moreira IZ, Jones SA, Lang T, Riccardi C, Teixeira MM, Harris J, Morand EF, Sousa LP. Glucocorticoid-induced leucine zipper modulates macrophage polarization and apoptotic cell clearance. Pharmacol Res 2020; 158:104842. [PMID: 32413484 DOI: 10.1016/j.phrs.2020.104842] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
Macrophages are professional phagocytes that display remarkable plasticity, with a range of phenotypes that can be broadly characterized by the M1/M2 dichotomy. Glucocorticoid (GC)-induced leucine zipper (GILZ) is a protein known to mediate anti-inflammatory and some pro-resolving actions, including as neutrophil apoptosis. However, the role of GILZ in key macrophage function is not well understood. Here, we investigated the role of GILZ on macrophage reprogramming and efferocytosis. Using murine bone-marrow-derived macrophages (BMDMs), we found that GILZ was expressed in naive BMDMs and exhibited increased expression in M2-like macrophages (IL4-differentiated). M1-like macrophages (IFN/LPS-differentiated) from GILZ-/- mice showed higher expression of the M1 markers CD86, MHC class II, iNOS, IL-6 and TNF-α, associated with increased levels of phosphorylated STAT1 and lower IL-10 levels, compared to M1-differentiated cells from WT mice. There were no changes in the M2 markers CD206 and arginase-1 in macrophages from GILZ-/- mice differentiated with IL-4, compared to cells from WT animals. Treatment of M1-like macrophages with TAT-GILZ, a cell-permeable GILZ fusion protein, decreased the levels of CD86 and MHC class II in M1-like macrophages without modifying CD206 levels in M2-like macrophages. In line with the in vitro data, increased numbers of M1-like macrophages were found into the pleural cavity of GILZ-/- mice after LPS-injection, compared to WT mice. Moreover, efferocytosis was defective in the context of GILZ deficiency, both in vitro and in vivo. Conversely, treatment of LPS-injected mice with TAT-GILZ promoted inflammation resolution, associated with lower numbers of M1-like macrophages and increased efferocytosis. Collectively, these data indicate that GILZ is a regulator of important macrophage functions, contributing to macrophage reprogramming and efferocytosis, both key steps for the resolution of inflammation.
Collapse
Affiliation(s)
- Juliana P Vago
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Rheumatology Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Victoria, Australia
| | - Izabela Galvão
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Graziele L Negreiros-Lima
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lívia C R Teixeira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kátia M Lima
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michelle A Sugimoto
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Z Moreira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sarah A Jones
- Rheumatology Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Victoria, Australia
| | - Tali Lang
- Rheumatology Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Victoria, Australia
| | - Carlo Riccardi
- Departament of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Mauro M Teixeira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - James Harris
- Rheumatology Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Victoria, Australia
| | - Eric F Morand
- Rheumatology Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Victoria, Australia
| | - Lirlândia P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
50
|
Gibson BH, Duvernay MT, Moore‐Lotridge SN, Flick MJ, Schoenecker JG. Plasminogen activation in the musculoskeletal acute phase response: Injury, repair, and disease. Res Pract Thromb Haemost 2020; 4:469-480. [PMID: 32548548 PMCID: PMC7293893 DOI: 10.1002/rth2.12355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 12/22/2022] Open
Abstract
The musculoskeletal system is critical for movement and the protection of organs. In addition to abrupt injuries, daily physical demands inflict minor injuries, necessitating a coordinated process of repair referred to as the acute-phase response (APR). Dysfunctional APRs caused by severe injuries or underlying chronic diseases are implicated in pathologic musculoskeletal repair, resulting in decreased mobility and chronic pain. The molecular mechanisms behind these phenomena are not well understood, hindering the development of clinical solutions. Recent studies indicate that, in addition to regulating intravascular clotting, the coagulation and fibrinolytic systems are also entrenched in tissue repair. Although plasmin and fibrin are considered antithetical to one another in the context of hemostasis, in a proper APR, they complement one another within a coordinated spatiotemporal framework. Once a wound is contained by fibrin, activation of plasmin promotes the removal of fibrin and stimulates angiogenesis, tissue remodeling, and tissue regeneration. Insufficient fibrin deposition or excessive plasmin-mediated fibrinolysis in early convalescence prevents injury containment, causing bleeding. Alternatively, excess fibrin deposition and/or inefficient plasmin activity later in convalescence impairs musculoskeletal repair, resulting in tissue fibrosis and osteoporosis, while inappropriate fibrin or plasmin activity in a synovial joint can cause arthritis. Together, these pathologic conditions lead to chronic pain, poor mobility, and diminished quality of life. In this review, we discuss both fibrin-dependent and -independent roles of plasminogen activation in the musculoskeletal APR, how dysregulation of these mechanisms promote musculoskeletal degeneration, and the possibility of therapeutically manipulating plasmin or fibrin to treat musculoskeletal disease.
Collapse
Affiliation(s)
| | - Matthew T. Duvernay
- Department of PharmacologyVanderbilt UniversityNashvilleTNUSA
- Department of OrthopaedicsVanderbilt University Medical CenterNashvilleTNUSA
- Center for Bone BiologyVanderbilt University Medical CenterNashvilleTNUSA
| | | | - Matthew J. Flick
- Department of Pathology and Laboratory MedicineUniversity of North Carolina‐Chapel HillNCUSA
- UNC Blood Research CenterChapel HillNCUSA
| | - Jonathan G. Schoenecker
- Department of PharmacologyVanderbilt UniversityNashvilleTNUSA
- Department of OrthopaedicsVanderbilt University Medical CenterNashvilleTNUSA
- Center for Bone BiologyVanderbilt University Medical CenterNashvilleTNUSA
- Department of Pathology, Microbiology, and ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
- Department of PediatricsVanderbilt University Medical CenterNashvilleTNUSA
| |
Collapse
|