1
|
Frostegård A, Haegerstrand A. New Therapeutic Strategies in Retinal Vascular Diseases: A Lipid Target, Phosphatidylserine, and Annexin A5-A Future Theranostic Pairing in Ophthalmology. Pharmaceuticals (Basel) 2024; 17:979. [PMID: 39204083 PMCID: PMC11357257 DOI: 10.3390/ph17080979] [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: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 09/03/2024] Open
Abstract
Despite progress in the management of patients with retinal vascular and degenerative diseases, there is still an unmet clinical need for safe and effective therapeutic options with novel mechanisms of action. Recent mechanistic insights into the pathogenesis of retinal diseases with a prominent vascular component, such as retinal vein occlusion (RVO), diabetic retinopathy (DR) and wet age-related macular degeneration (AMD), may open up new treatment paradigms that reach beyond the inhibition of vascular endothelial growth factor (VEGF). Phosphatidylserine (PS) is a novel lipid target that is linked to the pathophysiology of several human diseases, including retinal diseases. PS acts upstream of VEGF and complement signaling pathways. Annexin A5 is a protein that targets PS and inhibits PS signaling. This review explores the current understanding of the potential roles of PS as a target and Annexin A5 as a therapeutic. The clinical development status of Annexin A5 as a therapeutic and the potential utility of PS-Annexin A5 as a theranostic pairing in retinal vascular conditions in particular is described.
Collapse
Affiliation(s)
- Anna Frostegård
- Annexin Pharmaceuticals AB, Kammakargatan 48, S-111 60 Stockholm, Sweden
- Unit of Immunology and Chronic Disease, IMM, Karolinska Institute, S-171 77 Stockholm, Sweden
| | | |
Collapse
|
2
|
Dong Y, Jia Z, Kang B, Zhang W. Annexin-A5 monomer as a membrane repair agent for the treatment of renal ischemia-reperfusion injury. Mol Biol Rep 2024; 51:679. [PMID: 38796668 DOI: 10.1007/s11033-024-09606-2] [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: 02/23/2024] [Accepted: 05/01/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (IRI) is one of the causes of acute kidney injury. Annexin A5 (AnxA5), a calcium-dependent cell membrane-binding protein, shows protective effects in various organ IRI models. This study explored the therapeutic effect of exogenous AnxA5 monomer protein on renal IRI and its potential mechanism of action. METHODS AND RESULTS Different doses of AnxA5 were injected intravenously to treat bilateral renal IRI in SD rats. This model confirmed the protective effects of AnxA5 on kidney structure and function. In vitro, HK-2 cells were subjected to hypoxia for 12 h, followed by restoration of normal oxygen supply to simulate IRI. In vitro experiments demonstrated the mechanism of action of AnxA5 by measuring cellular activity and permeability. A comparison of the mutant AnxA5 protein M23 and the application of a calcium-free culture medium further validated the protective effect of AnxA5 by forming a network structure. CONCLUSIONS Exogenous AnxA5 monomers prevented renal IRI by binding to the damaged renal tubular epithelial cell membrane, forming a two-dimensional network structure to maintain cell membrane integrity, and ultimately prevent cell death.
Collapse
Affiliation(s)
- Yushan Dong
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhuoxuan Jia
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bijun Kang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
3
|
Zhang L, Zhao S, Wang Y. Diannexin alleviates myocardial ischemia-reperfusion injury by orchestrating cardiomyocyte oxidative damage, macrophage polarization and fibrotic process by TLR4-NF-kB-mediated inactivation of NLRP3 inflammasome. Int Immunopharmacol 2024; 130:111668. [PMID: 38417368 DOI: 10.1016/j.intimp.2024.111668] [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: 11/12/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 03/01/2024]
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a pathogenic mechanism of myocardial infarction and heart failure, constituting a major health concern globally. Diannexin is a homodimer of recombinant human annexin V and elicits important roles in several I/R injuries. Nevertheless, its function in MI/R remains elusive. Here, Diannexin alleviated simulated I/R (SI/R)-induced cardiomyocyte death and oxidative injury by increasing cell viability and inhibiting cell apoptosis, ROS, lactate dehydrogenase, malondialdehyde production and anti-oxidative SOD activity. Diannexin inhibited SI/R-induced expression of fibrotic protein collagen I and collagen III. Furthermore, Diannexin suppressed LPS-induced macrophage polarization towards pro-inflammatory M1-like phenotype and enhanced IL-4-evoked anti-inflammatory M2 polarization. Concomitantly, Diannexin inhibited SI/R exposure-induced macrophage polarization to M1 subtypes. Importantly, conditioned medium (CM) from SI/R-stimulated macrophages evoked cardiomyocyte apoptosis, which was reversed when cells were co-cultured with CM from Diannexin-treated macrophages under SI/R conditions. Mechanically, the activation of TLR4/NF-κB/NLRP3 inflammasome signaling in SI/R-treated cells was mitigated by Diannexin. Reactivating this pathway antagonized the protective effects of Diannexin on SI/R-induced cardiomyocyte oxidative injury, fibrotic protein expression and macrophage polarization and M1 macrophage-induced apoptosis of cardiomyocytes. In vivo, Diannexin alleviated abnormal cardiac structure, dysfunction and collagen position in MI/R mice. Additionally, Diannexin reduced M1-polarized and elevated M2-polarized macrophages in heart tissues at five days post-MI/R. The activation of TLR4/NF-κB/NLRP3 inflammasome pathway in MI/R mice was attenuated after Diannexin administration. Together, Diannexin may alleviate the development of MI/R injury by directly regulating cardiomyocyte oxidative injury, fibrotic potential and indirectly affecting macrophage polarization-mediated cardiomyocyte apoptosis, indicating a promising therapeutic strategy for MI/R.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Clinical Laboratory Medicine, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Songlin Zhao
- Department of Clinical Laboratory Medicine, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yaqi Wang
- Department of Clinical Laboratory Medicine, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
4
|
Guo H, Zhang Y, Chu Y, Yang S, Zhang J, Qiao R. Recombinant protein diannexin prevents preeclampsia-like symptoms in a pregnant mouse model via reducing the release of microparticles. Front Med 2022; 16:919-931. [PMID: 36331793 DOI: 10.1007/s11684-021-0918-6] [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: 07/05/2021] [Accepted: 12/21/2021] [Indexed: 11/06/2022]
Abstract
Preeclampsia (PE) is characterized by placenta-mediated pregnancy complication. The only effective treatment for PE is the delivery of the placenta. However, this treatment may cause preterm birth and neonatal death. Therefore, preventing PE is needed. The mechanism of PE involves abnormal placentation, which leads to the release of anti-angiogenic and inflammatory mediators into maternal circulation. These mediators contribute to systemic vascular dysfunction, inflammatory responses, and excessive thrombin generation. Microparticles (MPs) are reportedly involved in PE by promoting the thromboinflammatory response. This study describes a strategy to prevent PE by reducing MP release using the recombinant protein, diannexin. Results showed that the patients with PE had elevated MP number and procoagulant activity and increased NLRP3 inflammasome activation. Additionally, diannexin remarkably reduced the release of MPs from activated cells by binding to phosphatidylserine exposed on the surface of activated cells. Moreover, in vivo results showed that diannexin could prevent PE-like symptoms by decreasing MPs and NLRP3 inflammasome activation in pregnant mice. Furthermore, diannexin effectively inhibited trophoblast cell activation and NLRP3 inflammasome activation in vitro. These findings suggested that diannexin inhibited MP release and might be an effective therapeutic strategy for preventing PE.
Collapse
Affiliation(s)
- Han Guo
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yuncong Zhang
- Department of Clinical Laboratory, Peking University International Hospital, Beijing, 102206, China
| | - Yaxin Chu
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Shuo Yang
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Jie Zhang
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Rui Qiao
- Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
5
|
Abstract
The cytokine storm (CS) in hyperinflammation is characterized by high levels of cytokines, extreme activation of innate as well as adaptive immune cells and initiation of apoptosis. High levels of apoptotic cells overwhelm the proper recognition and removal system of these cells. Phosphatidylserine on the apoptotic cell surface, which normally provides a recognition signal for removal, becomes a target for hemostatic proteins and secretory phospholipase A2. The dysregulation of these normal pathways in hemostasis and the inflammasome result in a prothrombotic state, cellular death, and end-organ damage. In this review, we provide the argument that this imbalance in recognition and removal is a common denominator regardless of the inflammatory trigger. The complex reaction of the immune defense system in hyperinflammation leads to self-inflicted damage. This common endpoint may provide additional options to monitor the progression of the inflammatory syndrome, predict severity, and may add to possible treatment strategies.
Collapse
|
6
|
The Nephrotoxin Puromycin Aminonucleoside Induces Injury in Kidney Organoids Differentiated from Induced Pluripotent Stem Cells. Cells 2022; 11:cells11040635. [PMID: 35203286 PMCID: PMC8870209 DOI: 10.3390/cells11040635] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/29/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Kidney diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), which can progress to end stage renal disease (ESRD), are a worldwide health burden. Organ transplantation or kidney dialysis are the only effective available therapeutic tools. Therefore, in vitro models of kidney diseases and the development of prospective therapeutic options are urgently needed. Within the kidney, the glomeruli are involved in blood filtration and waste excretion and are easily affected by changing cellular conditions. Puromycin aminonucleoside (PAN) is a nephrotoxin, which can be employed to induce acute glomerular damage and to model glomerular disease. For this reason, we generated kidney organoids from three iPSC lines and treated these with PAN in order to induce kidney injury. Morphological observations revealed the disruption of glomerular and tubular structures within the kidney organoids upon PAN treatment, which were confirmed by transcriptome analyses. Subsequent analyses revealed an upregulation of immune response as well as inflammatory and cell-death-related processes. We conclude that the treatment of iPSC-derived kidney organoids with PAN induces kidney injury mediated by an intertwined network of inflammation, cytoskeletal re-arrangement, DNA damage, apoptosis and cell death. Furthermore, urine-stem-cell-derived kidney organoids can be used to model kidney-associated diseases and drug discovery.
Collapse
|
7
|
Mui L, Martin CM, Tschirhart BJ, Feng Q. Therapeutic Potential of Annexins in Sepsis and COVID-19. Front Pharmacol 2021; 12:735472. [PMID: 34566657 PMCID: PMC8458574 DOI: 10.3389/fphar.2021.735472] [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: 07/05/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Sepsis is a continuing problem in modern healthcare, with a relatively high prevalence, and a significant mortality rate worldwide. Currently, no specific anti-sepsis treatment exists despite decades of research on developing potential therapies. Annexins are molecules that show efficacy in preclinical models of sepsis but have not been investigated as a potential therapy in patients with sepsis. Human annexins play important roles in cell membrane dynamics, as well as mediation of systemic effects. Most notably, annexins are highly involved in anti-inflammatory processes, adaptive immunity, modulation of coagulation and fibrinolysis, as well as protective shielding of cells from phagocytosis. These discoveries led to the development of analogous peptides which mimic their physiological function, and investigation into the potential of using the annexins and their analogous peptides as therapeutic agents in conditions where inflammation and coagulation play a large role in the pathophysiology. In numerous studies, treatment with recombinant human annexins and annexin analogue peptides have consistently found positive outcomes in animal models of sepsis, myocardial infarction, and ischemia reperfusion injury. Annexins A1 and A5 improve organ function and reduce mortality in animal sepsis models, inhibit inflammatory processes, reduce inflammatory mediator release, and protect against ischemic injury. The mechanisms of action and demonstrated efficacy of annexins in animal models support development of annexins and their analogues for the treatment of sepsis. The effects of annexin A5 on inflammation and platelet activation may be particularly beneficial in disease caused by SARS-CoV-2 infection. Safety and efficacy of recombinant human annexin A5 are currently being studied in clinical trials in sepsis and severe COVID-19 patients.
Collapse
Affiliation(s)
- Louise Mui
- Division of Critical Care, Department of Medicine, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| | - Claudio M Martin
- Division of Critical Care, Department of Medicine, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada.,Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
| | - Brent J Tschirhart
- Department of Physiology and Pharmacology, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| | - Qingping Feng
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada.,Department of Physiology and Pharmacology, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| |
Collapse
|
8
|
Long-circulating XTEN864-annexin A5 fusion protein for phosphatidylserine-related therapeutic applications. Apoptosis 2021; 26:534-547. [PMID: 34405304 PMCID: PMC8370750 DOI: 10.1007/s10495-021-01686-w] [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] [Accepted: 08/03/2021] [Indexed: 01/21/2023]
Abstract
Annexin A5 (anxA5) is a marker for apoptosis, but has also therapeutic potential in cardiovascular diseases, cancer, and, due to apoptotic mimicry, against dangerous viruses, which is limited by the short blood circulation. An 864-amino-acid XTEN polypeptide was fused to anxA5. XTEN864-anxA5 was expressed in Escherichia coli and purified using XTEN as tag. XTEN864-anxA5 was coupled with DTPA and indium-111. After intravenous or subcutaneous injection of 111In-XTEN864-anxA5, mouse blood samples were collected for blood half-life determination and organ samples for biodistribution using a gamma counter. XTEN864-anxA5 was labeled with 6S-IDCC to confirm binding to apoptotic cells using flow cytometry. To demonstrate targeting of atherosclerotic plaques, XTEN864-anxA5 was labeled with MeCAT(Ho) and administered intravenously to atherosclerotic ApoE−/− mice. MeCAT(Ho)-XTEN864-anxA5 was detected together with MeCAT(Tm)-MAC-2 macrophage antibodies by imaging mass cytometry (CyTOF) of aortic root sections. The ability of anxA5 to bind apoptotic cells was not affected by XTEN864. The blood half-life of XTEN864-anxA5 was 13 h in mice after IV injection, markedly longer than the 7-min half-life of anxA5. 96 h after injection, highest amounts of XTEN864-anxA5 were found in liver, spleen, and kidney. XTEN864-anxA5 was found to target the adventitia adjacent to atherosclerotic plaques. XTEN864-anxA5 is a long-circulating fusion protein that can be efficiently produced in E. coli and potentially circulates in humans for several days, making it a promising therapeutic drug.
Collapse
|
9
|
Kuypers FA, Rostad CA, Anderson EJ, Chahroudi A, Jaggi P, Wrammert J, Mantus G, Basu R, Harris F, Hanberry B, Camacho-Gonzalez A, Manoranjithan S, Vos M, Brown LA, Morris CR. Secretory phospholipase A2 in SARS-CoV-2 infection and multisystem inflammatory syndrome in children (MIS-C). Exp Biol Med (Maywood) 2021; 246:2543-2552. [PMID: 34255566 PMCID: PMC8649422 DOI: 10.1177/15353702211028560] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Secretory phospholipase 2 (sPLA2) acts as a mediator between proximal and distal events of the inflammatory cascade. Its role in SARS-CoV-2 infection is unknown, but could contribute to COVID-19 inflammasome activation and cellular damage. We present the first report of plasma sPLA2 levels in adults and children with COVID-19 compared with controls. Currently asymptomatic adults with a history of recent COVID-19 infection (≥4 weeks before) identified by SARS-CoV-2 IgG antibodies had sPLA2 levels similar to those who were seronegative (9 ± 6 vs.17 ± 28 ng/mL, P = 0.26). In contrast, children hospitalized with severe COVID-19 had significantly elevated sPLA2 compared with those with mild or asymptomatic SARS-CoV-2 infection (269 ± 137 vs. 2 ± 3 ng/mL, P = 0.01). Among children hospitalized with multisystem inflammatory syndrome in children (MIS-C), all had severe disease requiring pediatric intensive care unit (PICU) admission. sPLA2 levels were significantly higher in those with acute illness <10 days versus convalescent disease ≥10 days (540 ± 510 vs. 2 ± 1, P = 0.04). Thus, sPLA2 levels correlated with COVID-19 severity and acute MIS-C in children, implicating a role in inflammasome activation and disease pathogenesis. sPLA2 may be a useful biomarker to stratify risk and guide patient management for children with acute COVID-19 and MIS-C. Therapeutic compounds targeting sPLA2 and inflammasome activation warrant consideration.
Collapse
Affiliation(s)
- Frans A Kuypers
- Division of Hematology, Department of Pediatrics, University of California, San Francisco, CA 94609, USA
| | - Christina A Rostad
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Evan J Anderson
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA.,Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ann Chahroudi
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Preeti Jaggi
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Jens Wrammert
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Grace Mantus
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Rajit Basu
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Frank Harris
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Bradley Hanberry
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Andres Camacho-Gonzalez
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | | | - Miriam Vos
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Clinical and Translational Research, Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Lou Ann Brown
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Claudia R Morris
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Clinical and Translational Research, Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Plasma microparticles of sickle patients during crisis or taking hydroxyurea modify endothelium inflammatory properties. Blood 2021; 136:247-256. [PMID: 32285120 DOI: 10.1182/blood.2020004853] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/05/2020] [Indexed: 12/29/2022] Open
Abstract
Microparticles (MPs) are submicron extracellular vesicles exposing phosphatidylserine (PS), detected at high concentration in the circulation of sickle cell anemia (SS) patients. Several groups studied the biological effects of MPs generated ex vivo. Here, we analyzed for the first time the impact of circulating MPs on endothelial cells (ECs) from 60 sickle cell disease (SCD) patients. MPs were collected from SCD patients and compared with MPs isolated from healthy individuals (AA). Other plasma MPs were purified from SS patients before and 2 years after the onset of hydroxyurea (HU) treatment or during a vaso-occlusive crisis and at steady-state. Compared with AA MPs, SS MPs increased EC ICAM-1 messenger RNA and protein levels, as well as neutrophil adhesion. We showed that ICAM-1 overexpression was primarily caused by MPs derived from erythrocytes, rather than from platelets, and that it was abolished by MP PS capping using annexin V. MPs from SS patients treated with HU were less efficient to induce a proinflammatory phenotype in ECs compared with MPs collected before therapy. In contrast, MPs released during crisis increased ICAM-1 and neutrophil adhesion levels, in a PS-dependent manner, compared with MPs collected at steady-state. Furthermore, neutrophil adhesion was abolished by a blocking anti-ICAM-1 antibody. Our study provides evidence that MPs play a key role in SCD pathophysiology by triggering a proinflammatory phenotype of ECs. We also uncover a new mode of action for HU and identify potential therapeutics: annexin V and anti-ICAM-1 antibodies.
Collapse
|
11
|
Cohan C, Beattie G, Brigode W, Yeung L, Miraflor E, Victorino GP. Protective Effect of Phosphatidylserine Blockade in Hemorrhagic Shock. J Surg Res 2019; 245:604-609. [PMID: 31499368 DOI: 10.1016/j.jss.2019.07.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/14/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Phosphatidylserine (PS) is a key cell membrane phospholipid normally maintained on the inner cell surface but externalizes to the outer surface in response to cellular stress. We hypothesized that PS exposure mediates organ dysfunction in hemorrhagic shock. Our aims were to evaluate PS blockade on (1) pulmonary, (2) renal, and (3) gut function, as well as (4) serum lysophosphatidic acid (LPA), an inflammatory mediator generated by PS externalization, as a possible mechanism mediating organ dysfunction. MATERIALS AND METHODS Rats were either (1) monitored for 130 min (controls, n = 3), (2) hemorrhaged then resuscitated (hemorrhage only group, n = 3), or (3) treated with Diannexin (DA), a PS blocking agent, followed by hemorrhage and resuscitation (DA + hemorrhage group, n = 4). Pulmonary dysfunction was assessed by arterial partial pressure of oxygen, renal dysfunction by serum creatinine, and gut dysfunction by mesenteric endothelial permeability (LP). LPA levels were measured in all groups. RESULTS Pulmonary: there was no difference in arterial partial pressure of oxygen between groups. Renal: after resuscitation, creatinine levels were lower after PS blockade with DA versus hemorrhage only group (P = 0.01). Gut: LP was decreased after PS blockade with DA versus hemorrhage only group (P < 0.01). Finally, LPA levels were also lower after PS blockade with DA versus the hemorrhage only group but higher than the control group (P < 0.01). CONCLUSIONS PS blockade with DA decreased renal and gut dysfunction associated with hemorrhagic shock and attenuated the magnitude of LPA generation. Our findings suggest potential for therapeutic targets in the future that could prevent organ dysfunction associated with hemorrhagic shock.
Collapse
Affiliation(s)
- Caitlin Cohan
- East Bay Department of Surgery, University of California San Francisco, Oakland, California.
| | - Genna Beattie
- East Bay Department of Surgery, University of California San Francisco, Oakland, California
| | - William Brigode
- East Bay Department of Surgery, University of California San Francisco, Oakland, California
| | - Louise Yeung
- East Bay Department of Surgery, University of California San Francisco, Oakland, California
| | - Emily Miraflor
- East Bay Department of Surgery, University of California San Francisco, Oakland, California
| | - Gregory P Victorino
- East Bay Department of Surgery, University of California San Francisco, Oakland, California
| |
Collapse
|
12
|
Beattie G, Cohan C, Miraflor E, Brigode W, Victorino GP. Protective effect of phosphatidylserine blockade in sepsis induced organ dysfunction. Surgery 2019; 166:844-848. [PMID: 31285044 DOI: 10.1016/j.surg.2019.05.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Phosphatidylserine is usually an intracellularly oriented cell membrane phospholipid. Externalized phosphatidylserine on activated cells is a signal for phagocytosis. In sepsis, persistent phosphatidylserine exposure is also a signal for activation of the coagulation and inflammatory cascades. As such, phosphatidylserine may be a key molecule in sepsis induced cellular and organ injury. We hypothesize that phosphatidylserine blockade provides a protective effect in sepsis induced organ dysfunction. METHODS Sepsis was induced in adult female rats using an endotoxin model. Diannexin, a homodimer of annexin A5, was administered for phosphatidylserine blockade. Rats were allocated to control (n = 5), sepsis (n = 6), or sepsis and phosphatidylserine blockade (n = 9) groups. Gut, pulmonary, renal, and hematologic dysfunctions were evaluated by mesenteric microvascular fluid leak, partial pressure of oxygen, serum creatinine, activated clotting time, and glomerular fibrin deposition, respectively. RESULTS Rats in the sepsis group demonstrated gut, renal, and hematologic dysfunction. Phosphatidylserine blockade reversed signs of gut dysfunction and mesenteric microvascular leak (P < .01). In addition, phosphatidylserine blockade corrected systemic coagulopathy, as measured by activated clotting time (P = .03) and glomerular fibrin deposition (P = .008). There was no difference in renal dysfunction (P = .1) or pulmonary dysfunction in any of the groups (P = .6). CONCLUSION In sepsis, phosphatidylserine blockade had a protective effect on gut dysfunction and coagulopathy. Increased phosphatidylserine exposure may be a key mediator of organ dysfunction and coagulopathy during sepsis. These data may provide insights into novel treatment options for septic patients.
Collapse
Affiliation(s)
- Genna Beattie
- Department of Surgery, University of California San Francisco East Bay, Oakland, CA.
| | - Caitlin Cohan
- Department of Surgery, University of California San Francisco East Bay, Oakland, CA
| | - Emily Miraflor
- Department of Surgery, University of California San Francisco East Bay, Oakland, CA
| | - William Brigode
- Department of Surgery, University of California San Francisco East Bay, Oakland, CA
| | - Gregory P Victorino
- Department of Surgery, University of California San Francisco East Bay, Oakland, CA
| |
Collapse
|
13
|
Attenuation of endothelial phosphatidylserine exposure decreases ischemia-reperfusion induced changes in microvascular permeability. J Trauma Acute Care Surg 2019. [PMID: 29538229 DOI: 10.1097/ta.0000000000001891] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Translocation of phosphatidylserine from the inner leaflet to the outer leaflet of the endothelial membrane via phospholipid scramblase-1 (PLSCR1) is an apoptotic signal responsible for the loss of endothelial barrier integrity after ischemia-reperfusion injury (IRI). We hypothesized that inhibiting phosphatidylserine expression on endothelial cells would attenuate IRI induced increases in hydraulic permeability (Lp). METHODS Mesenteric Lp was measured in rat post-capillary mesenteric venules subjected to IRI via superior mesenteric artery (SMA) occlusion (45 minutes) and release (300 minutes) in conjunction with several inhibitors of phosphatidylserine exposure as follows: (1) inhibition of PLSCR1 translocation (dithioerythritol, n = 3), (2) inhibition of PLSCR1 membrane trafficking (2-bromopalmitate [2-BP], n = 3), and (3) inhibition of ion exchange necessary for PLSCR1 function (4,4'-Diisothiocyano-2,2'-stilbenedisulfonic acid [DIDS], n = 3). Under the same IRI conditions, rats were also administered targeted inhibitors of phosphatidylserine exposure including knockdown of PLSCR1 (n = 3) using RNA interference (RNAi), and as a potential therapeutic tool Diannexin, a selective phosphatidylserine blocker (n = 3). RESULTS During IRI net Lp increased by 80% (p < 0.01). Net reductions of Lp were accomplished by 2-BP (46% reduction, p = 0.005), combined DET + 2-BP + DIDS (32% reduction, p = 0.04), RNAi (55% reduction, p = 0.002), Diannexin administered pre-SMA artery occlusion (73% reduction, p = 0.001), and post-SMA occlusion (70% reduction, p = 0.002). CONCLUSION Phosphatidylserine exposure is a key event in the pathogenesis of microvascular dysfunction during IRI. Clinically, inhibition of phosphatidylserine exposure is a promising strategy that may 1 day be used to mitigate the effects of IRI.
Collapse
|
14
|
Bern MM. Extracellular vesicles: how they interact with endothelium, potentially contributing to metastatic cancer cell implants. Clin Transl Med 2017; 6:33. [PMID: 28933058 PMCID: PMC5607152 DOI: 10.1186/s40169-017-0165-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/13/2017] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EV) are blebs of cellular membranes, which entrap small portions of subjacent cytosol. They are released from a variety of cells, circulate in the blood for an unknown length of time and come to rest on endothelial surfaces. They contribute to an array of physiologic pathways, the complexity of which is still being investigated. They contribute to metastatic malignant cell implants and tumor-related angiogenesis, possibly abetted by the tissue factor that they carry. It is thought that the adherence of the EV to endothelium is dependent upon a combination of their P-selectin glycoprotein ligand-1 and exposed phosphatidylserine, the latter of which is normally hidden on the inner bilayer of the intact cellular membrane. This manuscript reviews what is known about EV origins, their clearance from the circulation and how they contribute to malignant cell implants upon endothelium surfaces and subsequent tumor growth.
Collapse
Affiliation(s)
- Murray M Bern
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud, Albuquerque, NM, 87131, USA.
| |
Collapse
|
15
|
Effects of Remote Ischemic Preconditioning on Heme Oxygenase-1 Expression and Cutaneous Wound Repair. Int J Mol Sci 2017; 18:ijms18020438. [PMID: 28218659 PMCID: PMC5343972 DOI: 10.3390/ijms18020438] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 12/26/2022] Open
Abstract
Skin wounds may lead to scar formation and impaired functionality. Remote ischemic preconditioning (RIPC) can induce the anti-inflammatory enzyme heme oxygenase-1 (HO-1) and protect against tissue injury. We aim to improve cutaneous wound repair by RIPC treatment via induction of HO-1. RIPC was applied to HO-1-luc transgenic mice and HO-1 promoter activity and mRNA expression in skin and several other organs were determined in real-time. In parallel, RIPC was applied directly or 24h prior to excisional wounding in mice to investigate the early and late protective effects of RIPC on cutaneous wound repair, respectively. HO-1 promoter activity was significantly induced on the dorsal side and locally in the kidneys following RIPC treatment. Next, we investigated the origin of this RIPC-induced HO-1 promoter activity and demonstrated increased mRNA in the ligated muscle, heart and kidneys, but not in the skin. RIPC did not change HO-1 mRNA and protein levels in the wound 7 days after cutaneous injury. Both early and late RIPC did not accelerate wound closure nor affect collagen deposition. RIPC induces HO-1 expression in several organs, but not the skin, and did not improve excisional wound repair, suggesting that the skin is insensitive to RIPC-mediated protection.
Collapse
|
16
|
Shaver CM, Ware LB. Primary graft dysfunction: pathophysiology to guide new preventive therapies. Expert Rev Respir Med 2017; 11:119-128. [PMID: 28074663 DOI: 10.1080/17476348.2017.1280398] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Primary graft dysfunction (PGD) is a common complication of lung transplantation characterized by acute pulmonary edema associated with bilateral pulmonary infiltrates and hypoxemia in the first 3 post-operative days. Development of PGD is a predictor of poor short- and long-term outcomes after lung transplantation, but there are currently limited tools to prevent its occurrence. Areas covered: Several potentially modifiable donor, recipient, and operative risk factors for PGD have been identified. In addition, basic and translational studies in animals and ex vivo lung perfusion systems have identified several biomarkers and mechanisms of injury in PGD. In this review, we outline the clinical and genetic risk factors for PGD and summarize experimental data exploring PGD mechanisms, with a focus on strategies to reduce PGD risk and on potential novel molecular targets for PGD prevention. Expert commentary: Because of the clinical importance of PGD, development of new therapies for prevention and treatment is critically important. Improved understanding of the pathophysiology of clinical PGD provides a framework to explore novel agents to prevent or reverse PGD. Ex vivo lung perfusion provides a new opportunity for rapid development of therapeutics that target this devastating complication of lung transplantation.
Collapse
Affiliation(s)
- Ciara M Shaver
- a Department of Medicine , Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center , Nashville , TN , USA
| | - Lorraine B Ware
- a Department of Medicine , Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center , Nashville , TN , USA.,b Department of Pathology, Microbiology and Immunology , Vanderbilt University Medical Center , Nashville , TN , USA
| |
Collapse
|
17
|
Combes V, Latham SL, Wen B, Allison AC, Grau GER. DIANNEXIN DOWN-MODULATES TNF-INDUCED ENDOTHELIAL MICROPARTICLE RELEASE BY BLOCKING MEMBRANE BUDDING PROCESS. INTERNATIONAL JOURNAL OF INNOVATIVE MEDICINE AND HEALTH SCIENCE 2016; 7:1-11. [PMID: 28149531 PMCID: PMC5279986 DOI: 10.20530/ijimhs_7_1-11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Microparticles are now recognised as true biological effectors with a role in immunopathology through their ability to disseminate functional properties. Diannexin, a homodimer of annexin V, binds to PS with a higher affinity and longer blood half-life than the monomer, inhibits prothrombinase complex activity thereby diminishing coagulation and reperfusion injury mediators and prevent microvesicle-mediated material transfer. Our aim was to determine if Diannexin could modulate microparticle production by endothelial cells by interacting with the phosphatidylserine exposure occurring during the release of these vesicles. RESULTS In this study we showed that fluorescently labelled Diannexin binds to calcimycin-activated endothelial cells but not to resting cells. After overnight incubation, Diannexin enters cells and their released MP carry Diannexin. Some Diannexin seems to be processed via early endosomes and later is found in lysosomes. Both unlabelled Diannexin and fluorescent Diannexin inhibit MP release from TNF-activated endothelial cells. However, Diannexin treatment does not prevent endothelial activation by TNF. In addition, the inhibitory effect of Diannexin on MP release could be observed when cells were pre-, concomitantly or post-treated with cytokines. Scanning electron microscopy showed differences in the numbers and types of protuberances at the cell surface when cells were treated or not with Diannexin. Finally, there is no apparent congruency between fluorescent Diannexin labelling and surface protuberances as shown by correlative microscopy. CONCLUSIONS Altogether these data suggest that Diannexin can inhibit endothelial vesiculation by binding PS present either at the cell surface or at the level of the inner leaflet of the plasma membrane.
Collapse
Affiliation(s)
- Valéry Combes
- Vascular Immunology Unit, Faculty of Medicine & Bosch Institute, The University of Sydney, Medical Foundation Building, 92-94 Parramatta Road, Camperdown NSW 2006, Australia
- School of Life Sciences, The University of Technology, Sydney, Ultimo NSW 2007, Australia
| | - Sharissa L. Latham
- Vascular Immunology Unit, Faculty of Medicine & Bosch Institute, The University of Sydney, Medical Foundation Building, 92-94 Parramatta Road, Camperdown NSW 2006, Australia
| | - Beryl Wen
- Vascular Immunology Unit, Faculty of Medicine & Bosch Institute, The University of Sydney, Medical Foundation Building, 92-94 Parramatta Road, Camperdown NSW 2006, Australia
| | | | - Georges E. R. Grau
- Vascular Immunology Unit, Faculty of Medicine & Bosch Institute, The University of Sydney, Medical Foundation Building, 92-94 Parramatta Road, Camperdown NSW 2006, Australia
| |
Collapse
|
18
|
Cremers NAJ, Suttorp M, Gerritsen MM, Wong RJ, van Run-van Breda C, van Dam GM, Brouwer KM, Kuijpers-Jagtman AM, Carels CEL, Lundvig DMS, Wagener FADTG. Mechanical Stress Changes the Complex Interplay Between HO-1, Inflammation and Fibrosis, During Excisional Wound Repair. Front Med (Lausanne) 2015; 2:86. [PMID: 26697429 PMCID: PMC4678194 DOI: 10.3389/fmed.2015.00086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/24/2015] [Indexed: 01/11/2023] Open
Abstract
Mechanical stress following surgery or injury can promote pathological wound healing and fibrosis, and lead to functional loss and esthetic problems. Splinted excisional wounds can be used as a model for inducing mechanical stress. The cytoprotective enzyme heme oxygenase-1 (HO-1) is thought to orchestrate the defense against inflammatory and oxidative insults that drive fibrosis. Here, we investigated the activation of the HO-1 system in a splinted and non-splinted full-thickness excisional wound model using HO-1-luc transgenic mice. Effects of splinting on wound closure, HO-1 promoter activity, and markers of inflammation and fibrosis were assessed. After seven days, splinted wounds were more than three times larger than non-splinted wounds, demonstrating a delay in wound closure. HO-1 promoter activity rapidly decreased following removal of the (epi)dermis, but was induced in both splinted and non-splinted wounds during skin repair. Splinting induced more HO-1 gene expression in 7-day wounds; however, HO-1 protein expression remained lower in the epidermis, likely due to lower numbers of keratinocytes in the re-epithelialization tissue. Higher numbers of F4/80-positive macrophages, αSMA-positive myofibroblasts, and increased levels of the inflammatory genes IL-1β, TNF-α, and COX-2 were present in 7-day splinted wounds. Surprisingly, mRNA expression of newly formed collagen (type III) was lower in 7-day wounds after splinting, whereas, VEGF and MMP-9 were increased. In summary, these data demonstrate that splinting delays cutaneous wound closure and HO-1 protein induction. The pro-inflammatory environment following splinting may facilitate higher myofibroblast numbers and increase the risk of fibrosis and scar formation. Therefore, inducing HO-1 activity against mechanical stress-induced inflammation and fibrosis may be an interesting strategy to prevent negative effects of surgery on growth and function in patients with orofacial clefts or in patients with burns.
Collapse
Affiliation(s)
- Niels A J Cremers
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands ; Experimental Rheumatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Maarten Suttorp
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Marlous M Gerritsen
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine , Stanford, CA , USA
| | - Coby van Run-van Breda
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Gooitzen M van Dam
- Department of Surgery, University Medical Center Groningen , Groningen , Netherlands
| | - Katrien M Brouwer
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, MOVE Research Institute Amsterdam , Amsterdam , Netherlands ; Association of Dutch Burn Centers , Beverwijk , Netherlands
| | - Anne Marie Kuijpers-Jagtman
- Department of Orthodontics and Craniofacial Biology, Cleft Palate Craniofacial Center, Radboud University Medical Center , Nijmegen , Netherlands
| | - Carine E L Carels
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Ditte M S Lundvig
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Frank A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| |
Collapse
|
19
|
Hashimoto K, Kim H, Oishi H, Chen M, Iskender I, Sakamoto J, Ohsumi A, Guan Z, Hwang D, Waddell TK, Cypel M, Liu M, Keshavjee S. Annexin V homodimer protects against ischemia reperfusion-induced acute lung injury in lung transplantation. J Thorac Cardiovasc Surg 2015; 151:861-869. [PMID: 26725713 DOI: 10.1016/j.jtcvs.2015.10.112] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/01/2015] [Accepted: 10/18/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE We hypothesized that administration of a homodimer of recombinant annexin V, diannexin, could shield phosphatidylserine on the endothelium, and inhibit leukocyte and platelet adhesion, thereby potentially reducing ischemia reperfusion injury (IRI) in lung transplantation. This hypothesis was tested using a rat syngeneic single left-lung transplant model. METHODS Rats were randomly assigned to receive diannexin (DN group; n = 10) or normal saline (control group; n = 10). Diannexin (1000 μg/kg) was administered to the donor lung in the pulmonary flush solution, and to the recipient intravenously, 5 minutes after initiation of reperfusion. Grafts were reperfused for 2 hours. RESULTS The transplanted grafts in the DN group performed significantly better in gas exchange with higher partial pressure of oxygen (control group: 179 ± 121 vs DN group: 330 ± 54 mm Hg; P = .007) and lower partial pressure of carbon dioxide (control: 55.1 ± 26 vs DN: 34.2 ± 11 mm Hg; P = .04), as well as lower peak airway pressure (control: 20.5 ± 8.5 vs DN: 12.0 ± 7.9 cm H2O; P = .035) after 2 hours of reperfusion. Wet-to-dry lung weight ratio (P = .054), and alveolar fibrin deposition score (P = .04), were reduced in the DN group. Caspase-cleaved cytokeratin 18 in plasma (a marker of epithelial apoptosis) was significantly reduced in the DN group (P = .013). Furthermore, gene-expression levels of proinflammatory cytokines in the transplanted graft, including interleukin-6 (P = .04) and macrophage inflammatory protein 2 (P = .03) were significantly decreased in the DN group. CONCLUSIONS A homodimer of recombinant annexin V reduced ischemia reperfusion injury in a lung transplant animal model, by reducing cell death and tissue inflammation.
Collapse
Affiliation(s)
- Kohei Hashimoto
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hyunhee Kim
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hisashi Oishi
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Manyin Chen
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ilker Iskender
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Jin Sakamoto
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Akihiro Ohsumi
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Zehong Guan
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David Hwang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Thomas K Waddell
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
Salvadori M, Rosso G, Bertoni E. Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment. World J Transplant 2015; 5:52-67. [PMID: 26131407 PMCID: PMC4478600 DOI: 10.5500/wjt.v5.i2.52] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/16/2015] [Accepted: 04/27/2015] [Indexed: 02/05/2023] Open
Abstract
Ischemia/reperfusion injury is an unavoidable relevant consequence after kidney transplantation and influences short term as well as long-term graft outcome. Clinically ischemia/reperfusion injury is associated with delayed graft function, graft rejection, chronic rejection and chronic graft dysfunction. Ischemia/reperfusion affects many regulatory systems at the cellular level as well as in the renal tissue that result in a distinct inflammatory reaction of the kidney graft. Underlying factors of ischemia reperfusion include energy metabolism, cellular changes of the mitochondria and cellular membranes, initiation of different forms of cell death-like apoptosis and necrosis together with a recently discovered mixed form termed necroptosis. Chemokines and cytokines together with other factors promote the inflammatory response leading to activation of the innate immune system as well as the adaptive immune system. If the inflammatory reaction continues within the graft tissue, a progressive interstitial fibrosis develops that impacts long-term graft outcome. It is of particular importance in kidney transplantation to understand the underlying mechanisms and effects of ischemia/reperfusion on the graft as this knowledge also opens strategies to prevent or treat ischemia/reperfusion injury after transplantation in order to improve graft outcome.
Collapse
|
21
|
Dowlatshahi K, Ajami M, Pazoki-Toroudi H, Hajimiresmaiel SJ. ATP-dependent potassium channels are implicated in simvastatin pretreatment-induced inhibition of apoptotic cell death after renal ischemia/reperfusion injury. Med J Islam Repub Iran 2015; 29:191. [PMID: 26157709 PMCID: PMC4476217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/01/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Simvastatin is a widely used medication in cardiac care. Here we evaluate the role of ATP sensitive potassium (KATP) channels in simvastatin induced renal protection after renal ischemia/reperfusion (I/R) injury. METHODS A total of 81 male Wistar rats, were treated with simvastatin (10 and 20mg/kg/day; gavage, one week). Some groups received glibenclamide (KATP channel inhibitor; 5mg/kg) before ischemia (45min) and reperfusion (24h). Finally the kidneys were processed for histological analysis and measurement of biochemical parameters including tissue malondialdehyde (MDA), blood urea nitrogen (BUN), fractional excretion of sodium (FENa), creatinine clearance rate (CCr) and Bcl2-associated X protein (Bax) expression. RESULTS IR significantly increased serum Cr (p< 0.01) and BUN levels (p< 0.01), elevated FENa (p<0.01) and tissue MDA (p<0.01), and decreased CCr (p< 0.01) and induced histological damage. Bax pro-apoptotic protein was upregulated in renal tissue after I/R injury and downregulated in simvastatin pretreated group. Simvastatin at doses of 10 and 20mg/kg/day significantly reduced serum Cr and BUN levels (p< 0.05 vs. IR group), tissue MDA contents and FENa (p< 0.05 vs. I/R) and increased CCr (p< 0.05 vs. IR). Renal tissue injury was improved only in simvastatin 20mg/kg/day group (p< 0.05). Glibenclamide significantly abolished protective effects of simvastatin and increased serum Cr and BUN and FENa and decreased CCr (p< 0.05). It also abolished the effects of simvastatin on tissue injury and MDA contents and downregulated the Bax protein after IR injury (p< 0.05). CONCLUSION Opening of KATP channels is essential for simvastatin-induced renal protection against I/R injury.
Collapse
Affiliation(s)
- Kamran Dowlatshahi
- 1 Assistant Professor, Islamic Azad University, Najafabad Branch, Najafabad, Isfahan, Iran.
| | - Marjan Ajami
- 2 Assistant Professor, National Nutrition and Food Technology Research Institute, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamidreza Pazoki-Toroudi
- 3 Assistant Professor, Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
22
|
Murakami Y, Tian L, Voss OH, Margulies DH, Krzewski K, Coligan JE. CD300b regulates the phagocytosis of apoptotic cells via phosphatidylserine recognition. Cell Death Differ 2014; 21:1746-57. [PMID: 25034781 DOI: 10.1038/cdd.2014.86] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/14/2014] [Accepted: 05/21/2014] [Indexed: 01/15/2023] Open
Abstract
The CD300 receptor family members are a group of molecules that modulate a variety of immune cell processes. We show that mouse CD300b (CLM7/LMIR5), expressed on myeloid cells, recognizes outer membrane-exposed phosphatidylserine (PS) and does not, as previously reported, directly recognize TIM1 or TIM4. CD300b accumulates in phagocytic cups along with F-actin at apoptotic cell contacts, thereby facilitating their engulfment. The CD300b-mediated activation signal is conveyed through CD300b association with the adaptor molecule DAP12, and requires a functional DAP12 ITAM motif. Binding of apoptotic cells promotes the activation of the PI3K-Akt kinase pathway in macrophages, while silencing of CD300b expression diminishes PI3K-Akt kinase activation and impairs efferocytosis. Collectively, our data show that CD300b recognizes PS as a ligand, and regulates the phagocytosis of apoptotic cells via the DAP12 signaling pathway.
Collapse
Affiliation(s)
- Y Murakami
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - L Tian
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - O H Voss
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - D H Margulies
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - K Krzewski
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - J E Coligan
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Acute kidney injury (AKI) in transplant recipients is a prevalent condition with a broad list of potential inciting causes. This review highlights recent data describing the epidemiology and long-term consequences of transplant AKI, novel interventions in the management of delayed graft function (DGF), and noninvasive diagnostic strategies. RECENT FINDINGS The incidence and outcomes of nontransplant AKI are well documented, and similar data are emerging in the transplant setting with recent reports suggesting a high incidence rate and significant impact on long-term graft outcomes. DGF represents a 'pure' form of transplant AKI, and many interventional trials aiming to limit ischemia-reperfusion-induced injury have recently been reported or are currently ongoing. The search for accurate noninvasive predictors of DGF and acute rejection is ongoing and recent literature describes novel plasma and urine-based biomarkers as well as transcriptional profiling methods with high potential for clinical applicability. SUMMARY AKI in transplant recipients is a frequent occurrence with significant potential for poor long-term graft outcomes. Recent efforts to limit ischemia-reperfusion injury and diagnose transplant AKI via noninvasive methods may help to minimize the impact of AKI on future graft function.
Collapse
|
24
|
Kuypers FA. Hemoglobin S Polymerization and Red Cell Membrane Changes. Hematol Oncol Clin North Am 2014; 28:155-79. [DOI: 10.1016/j.hoc.2013.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
25
|
Winslow V, Vaivoda R, Vasilyev A, Dombkowski D, Douaidy K, Stark C, Drake J, Guilliams E, Choudhary D, Preffer F, Stoilov I, Christmas P. Altered leukotriene B4 metabolism in CYP4F18-deficient mice does not impact inflammation following renal ischemia. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:868-79. [PMID: 24632148 DOI: 10.1016/j.bbalip.2014.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 02/18/2014] [Accepted: 03/05/2014] [Indexed: 01/26/2023]
Abstract
Inflammatory responses to infection and injury must be restrained and negatively regulated to minimize damage to host tissue. One proposed mechanism involves enzymatic inactivation of the pro-inflammatory mediator leukotriene B4, but it is difficult to dissect the roles of various metabolic enzymes and pathways. A primary candidate for a regulatory pathway is omega oxidation of leukotriene B4 in neutrophils, presumptively by CYP4F3A in humans and CYP4F18 in mice. This pathway generates ω, ω-1, and ω-2 hydroxylated products of leukotriene B4, depending on species. We created mouse models targeting exons 8 and 9 of the Cyp4f18 allele that allows both conventional and conditional knockouts of Cyp4f18. Neutrophils from wild-type mice convert leukotriene B4 to 19-hydroxy leukotriene B4, and to a lesser extent 18-hydroxy leukotriene B4, whereas these products were not detected in neutrophils from conventional Cyp4f18 knockouts. A mouse model of renal ischemia-reperfusion injury was used to investigate the consequences of loss of CYP4F18 in vivo. There were no significant changes in infiltration of neutrophils and other leukocytes into kidney tissue as determined by flow cytometry and immunohistochemistry, or renal injury as assessed by histological scoring and measurement of blood urea nitrogen. It is concluded that CYP4F18 is necessary for omega oxidation of leukotriene B4 in neutrophils, and is not compensated by other CYP enzymes, but loss of this metabolic pathway is not sufficient to impact inflammation and injury following renal ischemia-reperfusion in mice.
Collapse
Affiliation(s)
- Valeria Winslow
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Rachel Vaivoda
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Aleksandr Vasilyev
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - David Dombkowski
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Karim Douaidy
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Christopher Stark
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Justin Drake
- Biology Department, Radford University, Radford, VA 24142, USA
| | - Evin Guilliams
- Biology Department, Radford University, Radford, VA 24142, USA
| | - Dharamainder Choudhary
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Frederic Preffer
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ivaylo Stoilov
- Department of Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Peter Christmas
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA; Biology Department, Radford University, Radford, VA 24142, USA.
| |
Collapse
|
26
|
Eid L, Tuchman S, Moudgil A. Late acute rejection: incidence, risk factors, and effect on graft survival and function. Pediatr Transplant 2014; 18:155-62. [PMID: 24372967 DOI: 10.1111/petr.12203] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/05/2013] [Indexed: 02/01/2023]
Abstract
Long-term graft survival and function has not kept pace with short-term success in kidney transplant (Tx) recipients. LAR ≥6 months post-Tx may contribute to lack of improvement; risk factors for LAR are not well known. Of 64 Tx recipients followed over six yr, 23 (35.9%) had LAR (LAR group) and 41 had no LAR (no LAR group). Of all variables, significant risk factors for LAR included DGF, (43.4% LAR vs. 14.6% in no LAR group, p = 0.0096); de novo DSA (65.2% vs. 26.8%, p = 0.003); mean COV% of TAC (41.8% vs. 34.6%, p = 0.03); and non-adherence (34.8% vs. 7.3%, p = 0.0043). DGF and DSA remained statistically significant (p = 0.002 and 0.003, respectively); COV% TAC had borderline significance (p = 0.057), and non-adherence was not significant on multivariate regression analysis. Patients with LAR had inferior graft survival and function, whereas graft function was stable in the no LAR group over a mean follow-up of 31.2 months. Patients with de novo DSA and DGF should be considered at risk of LAR; an early diagnosis and treatment of LAR may improve graft survival and function.
Collapse
Affiliation(s)
- Loai Eid
- Department of Nephrology, Children's National Medical Center, Washington, DC, USA
| | | | | |
Collapse
|
27
|
Grigoryev DN, Cheranova DI, Heruth DP, Huang P, Zhang LQ, Rabb H, Ye SQ. Meta-analysis of molecular response of kidney to ischemia reperfusion injury for the identification of new candidate genes. BMC Nephrol 2013; 14:231. [PMID: 24152794 PMCID: PMC4016589 DOI: 10.1186/1471-2369-14-231] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 10/11/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Accumulated to-date microarray data on ischemia reperfusion injury (IRI) of kidney represent a powerful source for identifying new targets and mechanisms of kidney IRI. In this study, we conducted a meta-analysis of gene expression profiles of kidney IRI in human, pig, rat, and mouse models, using a new scoring method to correct for the bias of overrepresented species. The gene expression profiles were obtained from the public repositories for 24 different models. After filtering against inclusion criteria 21 experimental settings were selected for meta-analysis and were represented by 11 rat models, 6 mouse models, and 2 models each for pig and human, with a total of 150 samples. Meta-analysis was conducted using expression-based genome-wide association study (eGWAS). The eGWAS results were corrected for a rodent species bias using a new weighted scoring algorithm, which favors genes with unidirectional change in expression in all tested species. RESULTS Our meta-analysis corrected for a species bias, identified 46 upregulated and 1 downregulated genes, of which 26 (55%) were known to be associated with kidney IRI or kidney transplantation, including LCN2, CCL2, CXCL1, HMOX1, ICAM1, ANXA1, and TIMP1, which justified our approach. Pathway analysis of our candidates identified "Acute renal failure panel" as the most implicated pathway, which further validates our new method. Among new IRI candidates were 10 novel (<5 published reports related to kidney IRI) and 11 new candidates (0 reports related to kidney IRI) including the most prominent candidates ANXA2, CLDN4, and TYROBP. The cross-species expression pattern of these genes allowed us to generate three workable hypotheses of kidney IRI, one of which was confirmed by an additional study. CONCLUSIONS Our first in the field kidney IRI meta-analysis of 150 microarray samples, corrected for a species bias, identified 10 novel and 11 new candidate genes. Moreover, our new meta-analysis correction method improved gene candidate selection by identifying genes that are model and species independent, as a result, function of these genes can be directly extrapolated to the disease state in human and facilitate translation of potential diagnostic or therapeutic properties of these candidates to the bedside.
Collapse
Affiliation(s)
- Dmitry N Grigoryev
- Division of Experimental and Translational Genetics, Department of Pediatrics, Children's Mercy Hospitals and Clinics, University of Missouri School of Medicine, Kansas City, MO, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
van Gelder T, Baan C, Vincenti F, Mannon RB. Report of the second joint meeting of ESOT and AST: current pipelines in biotech and pharma. Transpl Int 2013; 26:938-48. [PMID: 23822608 DOI: 10.1111/tri.12140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/15/2013] [Accepted: 06/10/2013] [Indexed: 12/14/2022]
Abstract
Following the first joint meeting organized by the European (ESOT) and American (AST) Societies of Transplantation in 2010, a second joint meeting was held in Nice, France, on October 12-14, 2012 at the Palais de la Mediterannee. Co-chairs of the scientific advisory committee were Dr. Flavio Vincenti (AST) and Dr. Teun Van Gelder (ESOT). The goal was to discuss the key unmet needs in solid organ transplantation with the opportunity to interrelate current basic research efforts with clinical translation. Thus, the topic of this second meeting "Transformational therapies and diagnostics in transplantation" was devised and a summary of this meeting follows.
Collapse
|
29
|
Powell JT, Tsapepas DS, Martin ST, Hardy MA, Ratner LE. Managing renal transplant ischemia reperfusion injury: novel therapies in the pipeline. Clin Transplant 2013; 27:484-91. [PMID: 23614480 DOI: 10.1111/ctr.12121] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2013] [Indexed: 01/26/2023]
Abstract
Ischemia reperfusion injury (IRI) is an early, non-specific inflammatory response that follows perfusion of warm blood into a cold asanguinous organ following transplantation. The occurrence of IRI may have a pivotal impact on acute and long-term renal allograft function. Initially, IRI contributes to delayed graft function (DGF), a term typically defined as the need for dialysis within one wk after renal transplantation. DGF frequently leads to prolonged hospital stay, increased healthcare costs, and potentially worse prognosis. Strategies to prevent IRI have so far been fairly limited, poorly defined, inadequately studied, and mostly anecdotal. The purpose of this review is to summarize the existing and novel therapies, which may mitigate IRI in renal transplantation. Agents currently in the pipeline include: Diannexin, which reduces endothelial cell injury by shielding phosphatidylserine; YSPSL, which mimics the binding portion of P-selectin glycoprotein ligand-1 to competitively inhibit translocation of P-selectin and recruitment of polymorphonuclear leukocytes to the surface of endothelial cells; and I5NP, a synthetic small interfering ribonucleic acid that results in the inhibition of p53 expression. These agents represent an exciting frontier in transplant pharmacotherapy; they are in various phases of investigation and may have broader benefits in reducing complications of DGF.
Collapse
Affiliation(s)
- Jaclyn T Powell
- Department of Pharmacy, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA.
| | | | | | | | | |
Collapse
|
30
|
How systemic inflammation modulates adenosine metabolism and adenosine receptor expression in humans in vivo. Crit Care Med 2012; 40:2609-16. [DOI: 10.1097/ccm.0b013e318259205b] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
31
|
Rand ML, Wang H, Pluthero FG, Stafford AR, Ni R, Vaezzadeh N, Allison AC, Kahr WHA, Weitz JI, Gross PL. Diannexin, an annexin A5 homodimer, binds phosphatidylserine with high affinity and is a potent inhibitor of platelet-mediated events during thrombus formation. J Thromb Haemost 2012; 10:1109-19. [PMID: 22463102 DOI: 10.1111/j.1538-7836.2012.04716.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Shielding of procoagulant phosphatidylserine (PS) with annexin A5 attenuates thrombosis, but annexin A5 (35.7 kDa) is rapidly cleared from the circulation. In contrast, Diannexin, a 73.1 kDa homodimer of annexin A5, has an extended half-life. OBJECTIVES To quantify the affinity of Diannexin for PS, examine its interaction with activated platelets and determine its effects on platelet-mediated events during thrombus formation. METHODS The affinities of Diannexin and annexin A5 for PS-containing lipid bilayers were compared using surface plasmon resonance, and binding to activated platelets was assessed by flow cytometry. Calibrated automated thrombography and thromboelastography were employed to study the effects of Diannexin on thrombin generation and platelet-fibrin clot formation, respectively, whereas intravital videomicroscopy was used to examine its effect on platelet accumulation and activation after laser-induced injury to murine cremaster arterioles, and a tail tip bleeding model was used to explore its effects on hemostasis. RESULTS Diannexin and annexin A5 bind PS with K(D) values of 0.6 and 5 nm, respectively, and both bind to the same subpopulation of PS-exposing platelets. Diannexin inhibited thrombin generation and platelet-fibrin clot formation in vitro at 10 nm (P<0.05-0.001 compared with control), and reduced platelet accumulation at 1 μg g(-1) (P<0.05) and activation at 0.25 μg g(-1) (P<0.001) in experimentally induced arterial thrombi in mice while increasing blood loss at 1 μg g(-1) (P<0.01). CONCLUSIONS Diannexin binds to PS with high affinity and is a potent inhibitor of platelet-mediated events during thrombus formation.
Collapse
Affiliation(s)
- M L Rand
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Manavalan A, Feng L, Sze SK, Hu JM, Heese K. New insights into the brain protein metabolism of Gastrodia elata-treated rats by quantitative proteomics. J Proteomics 2012; 75:2468-79. [PMID: 22402058 DOI: 10.1016/j.jprot.2012.02.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 02/13/2012] [Accepted: 02/20/2012] [Indexed: 01/05/2023]
Abstract
Gastrodia elata (tianma) is a traditional Chinese herbal medicine (TCM) often used for the treatment of cerebrovascular diseases. In this study, we investigated the effects of tianma on the brain protein metabolism by quantitative proteomics to gain evidence for a direct relationship between tianma treatment and brain functions. One-year-old rats were treated with tianma (~2.5 g/kg/day) for 3months and the brain tissue proteome was analyzed by using the iTRAQ (isobaric tag for relative and absolute quantification) technology. According to our results, the long-term treatment with tianma could modulate the brain protein metabolism at the proteome level by down-regulating the expressions of various proteins, such as Gnao1 and Dctn2, which are related to neuronal growth cone control and synaptic activities. In addition, tianma treatment also induced the up-regulation of molecular chaperons and proteins related to the misfolded protein response, like Anxa5, and also other proteins involved in Huntington's disease (HD) (e.g. Pacsin1 and Arf3). Concluding, tianma could eventually contribute to activities related to synaptic plasticity and neuro-restorative processes and thus might be a novel candidate agent for the treatment of neurodegenerative diseases by regulating the brain proteome.
Collapse
Affiliation(s)
- Arulmani Manavalan
- School of Biological Sciences, College of Science, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | | | | | | | | |
Collapse
|
33
|
Ewing MM, Karper JC, Sampietro ML, de Vries MR, Pettersson K, Jukema JW, Quax PHA. Annexin A5 prevents post-interventional accelerated atherosclerosis development in a dose-dependent fashion in mice. Atherosclerosis 2012; 221:333-40. [PMID: 22341596 DOI: 10.1016/j.atherosclerosis.2012.01.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 01/17/2012] [Accepted: 01/24/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND Activated cells in atherosclerotic lesions expose phosphatidylserine (PS) on their surface. Annexin A5 (AnxA5) binds to PS and is used for imaging atherosclerotic lesions. Recently, AnxA5 was shown to inhibit vascular inflammatory processes after vein grafting. Here, we report a therapeutic role for AnxA5 in post-interventional vascular remodeling in a mouse model mimicking percutaneous coronary intervention (PCI). METHODS AND RESULTS Associations between the rs4833229 (OR = 1.29 (CI 95%), p(allelic) = 0.011) and rs6830321 (OR = 1.35 (CI 95%), p(allelic) = 0.003) SNPs in the AnxA5 gene and increased restenosis-risk in patients undergoing PCI were found in the GENDER study. To evaluate AnxA5 effects on post-interventional vascular remodeling and accelerated atherosclerosis development in vivo, hypercholesterolemic ApoE(-/-) mice underwent femoral arterial cuff placement to induce intimal thickening. Dose-dependent effects were investigated after 3 days (effects on inflammation and leukocyte recruitment) or 14 days (effects on remodeling) after cuff placement. Systemically administered AnxA5 in doses of 0.1, 0.3 and 1.0mg/kg compared to vehicle reduced early leukocyte and macrophage adherence up to 48.3% (p = 0.001) and diminished atherosclerosis development by 71.2% (p = 0.012) with a reduction in macrophage/foam cell presence. Moreover, it reduced the expression of the endoplasmic reticulum stress marker GRP78/BiP, indicating lower inflammatory activity of the cells present. CONCLUSIONS AnxA5 SNPs could serve as markers for restenosis after PCI and AnxA5 therapeutically prevents vascular remodeling in a dose-dependent fashion, together indicating clinical potential for AnxA5 against post-interventional remodeling.
Collapse
Affiliation(s)
- M M Ewing
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|