1
|
Salinas MD, Martínez CM, Roca FJ, García-Bernal D, Martínez-Morga M, Rodríguez-Madoz JR, Prósper F, Zapata AG, Moraleda JM, Martínez S, Valdor R. Chaperone-mediated autophagy sustains pericyte stemness necessary for brain tissue homeostasis. J Adv Res 2025:S2090-1232(25)00259-0. [PMID: 40286844 DOI: 10.1016/j.jare.2025.04.015] [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: 07/30/2024] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Pericytes (PCs) are mural cells exhibiting some mesenchymal stem cell (MSC) properties and contribute to tissue regeneration after injury. We have previously shown that glioblastoma cancer cells induce in PCs, a pathogenic upregulation of chaperone-mediated autophagy (CMA) which modulates immune functions and MSC-like properties to support tumor growth. OBJECTIVES The aim of the study was to interrogate the role of CMA-regulated MSC properties in PCs in the context of tissue repair during inflammation triggered by a demyelinating injury. METHODS Studies of RNA-seq were done PCs with (WT) and without (LAMP-2A KO) CMA. Cell characterization related to stemness, lineage and morphology was done in WT and KO PCs. Secretome analysis and cell differentiation assay using the supernatants from CMA-efficient and deficient PCs cultures was done in mesenchymal cells. Inflammatory response of brain cells was assessed with WT and KO PCs secretome. To corroborate in vitro results, CMA modulation in response to inflammation in PCs and tissue repair markers were measured in the lesion areas of a demyelination mouse model and correlated with the tissue reparation after intravenous PC administration. An inflammatory mediator was used to study effects on PC-CMA activity. RESULTS We found that inflammatory mediators such as IFNγ downregulate CMA in PCs, suppressing PC stemness and promoting a pro-inflammatory secretome. Restoration of PC CMA activity during inflammation maintains PC MSC properties and induces an MSC-like proteome which decreases inflammation and promotes tissue repair. We identified secreted proteins involved in regenerative and protective processes, and therefore, necessary to restore brain tissue homeostasis after inflammation induced by a demyelinating injury. CONCLUSION we show that manipulation of CMA activity in host PCs could be a useful therapeutical approach in the context of brain inflammation, which might be extended to other diseases where the pericyte has a key role in response to inflammation.
Collapse
Affiliation(s)
- María Dolores Salinas
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain
| | | | - Francisco J Roca
- Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Unit of Infectious Disease Pathology, Clinical Microbiology and Tropical Medicine, IMIB, 30120 Murcia, Spain
| | - David García-Bernal
- Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain; Virgen de la Arrixaca University Hospital, Hematopoietic Transplant Group, IMIB, 30120 Murcia, Spain
| | - Marta Martínez-Morga
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain
| | - Juan R Rodríguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Navarra, Spain; Centro de investigación Biomédica en Red de Cancer (CIBERONC), Madrid, Spain
| | - Felipe Prósper
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Navarra, Spain; Centro de investigación Biomédica en Red de Cancer (CIBERONC), Madrid, Spain; Department of Dermatology and Cell Therapy, Clinica Universidad de Navarra (CUN), IdiSNA, 31008 Pamplona Navarra, Spain; Cancer Center Clinica Universidad de Navarra (CCUN), 31008 Pamplona, Navarra, Spain
| | - Agustín G Zapata
- Department of Cell Biology, Faculty of Biology, Complutense University, 28040 Madrid, Spain
| | - Jose María Moraleda
- Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain; Virgen de la Arrixaca University Hospital, Hematopoietic Transplant Group, IMIB, 30120 Murcia, Spain
| | - Salvador Martínez
- Instituto de Neurociencias-Miguel Hernández University (UMH-CSIC), 03550, San Juan de Alicante, ISABIAL, CIBERSAM, Alicante, Spain
| | - Rut Valdor
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain.
| |
Collapse
|
2
|
Hudák A, Pusztai D, Letoha A, Letoha T. Mutual Inhibition of Antithrombin III and SARS-CoV-2 Cellular Attachment to Syndecans: Implications for COVID-19 Treatment and Vaccination. Int J Mol Sci 2024; 25:7534. [PMID: 39062776 PMCID: PMC11277105 DOI: 10.3390/ijms25147534] [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: 06/18/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Antithrombin III (ATIII) is a potent endogenous anticoagulant that binds to heparan sulfate proteoglycans (HSPGs) on endothelial cells' surfaces. Among these HSPGs, syndecans (SDCs) are crucial as transmembrane receptors bridging extracellular ligands with intracellular signaling pathways. Specifically, syndecan-4 (SDC4) has been identified as a key receptor on endothelial cells for transmitting the signaling effects of ATIII. Meanwhile, SDCs have been implicated in facilitating the cellular internalization of SARS-CoV-2. Given the complex interactions between ATIII and SDC4, our study analyzed the impact of ATIII on the virus entry into host cells. While ATIII binds to all SDC isoforms, it shows the strongest affinity for SDC4. SDCs' heparan sulfate chains primarily influence ATIII's SDC attachment, although other parts might also play a role in ATIII's dominant affinity toward SDC4. ATIII significantly reduces SARS-CoV-2's cellular entry into cell lines expressing SDCs, suggesting a competitive inhibition mechanism at the SDC binding sites, particularly SDC4. Conversely, the virus or its spike protein decreases the availability of SDCs on the cell surface, reducing ATIII's cellular attachment and hence contributing to a procoagulant environment characteristic of COVID-19.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (D.P.)
| | - Dávid Pusztai
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (D.P.)
| | - Annamária Letoha
- Department of Medicine, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Tamás Letoha
- Pharmacoidea Ltd., 6726 Szeged, Hungary; (A.H.); (D.P.)
| |
Collapse
|
3
|
Kraaijvanger R, Janssen Bonás M, Grutters JC, Paspali I, Veltkamp M, de Kleijn DPV, van Moorsel CHM. Decreased serpin C1 in extracellular vesicles predicts response to methotrexate treatment in patients with pulmonary sarcoidosis. Respir Res 2024; 25:166. [PMID: 38627696 PMCID: PMC11020913 DOI: 10.1186/s12931-024-02809-y] [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: 11/03/2023] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Sarcoidosis is a systemic granulomatous disease of unknown etiology primarily affecting the lungs. Treatment is needed when disease symptoms worsen and organ function deteriorates. In pulmonary sarcoidosis, prednisone and methotrexate (MTX) are the most common anti-inflammatory therapies. However, there is large inter-patient variability in response to treatment, and predictive response markers are currently lacking. OBJECTIVE In this study, we investigated the predictive potential of biomarkers in extracellular vesicles (EVs) isolated from biobanked serum of patients with pulmonary sarcoidosis stored prior to start of therapy. METHODS Protein concentrations of a four-protein test panel of inflammatory proteins were measured in a discovery (n = 16) and replication (n = 129) cohort of patients with sarcoidosis and 47 healthy controls. Response to therapy was defined as an improvement of the absolute score of > 5% forced vital capacity (FVC) and/or > 10% diffusion lung of carbon monoxide (DLCO) after 24 weeks compared to baseline (before treatment). RESULTS Serum protein levels differed between EV fractions and serum, and between sarcoidosis cases and controls. Serpin C1 concentrations in the low density lipid particle EV fraction were lower at baseline in the group of patients with a good response to MTX treatment in both the discovery cohort (p = 0.059) and in the replication cohort (p = 0.032). EV Serpin C1 showed to be a significant predictor for response to treatment with MTX (OR 0.4; p = 0.032). CONCLUSION This study shows that proteins isolated from EVs harbor a distinct signal and have potential as new predictive therapy response biomarkers in sarcoidosis.
Collapse
Affiliation(s)
- Raisa Kraaijvanger
- Department of Pulmonology, St Antonius Hospital, Interstitial Lung Diseases Center of Excellence, Nieuwegein, The Netherlands
| | - Montse Janssen Bonás
- Department of Pulmonology, St Antonius Hospital, Interstitial Lung Diseases Center of Excellence, Nieuwegein, The Netherlands
| | - Jan C Grutters
- Department of Pulmonology, St Antonius Hospital, Interstitial Lung Diseases Center of Excellence, Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Center, Utrecht, The Netherlands
| | - Ioanna Paspali
- Department of Vascular Surgery, University Medical Center, Utrecht, The Netherlands
| | - Marcel Veltkamp
- Department of Pulmonology, St Antonius Hospital, Interstitial Lung Diseases Center of Excellence, Nieuwegein, The Netherlands
- Division of Heart and Lungs, University Medical Center, Utrecht, The Netherlands
| | | | - Coline H M van Moorsel
- Department of Pulmonology, St Antonius Hospital, Interstitial Lung Diseases Center of Excellence, Nieuwegein, The Netherlands.
| |
Collapse
|
4
|
Miura T, Okuda T, Suzuki K, Okada H, Tomita H, Takada C, Mori K, Asano H, Kano S, Wakayama Y, Fukuda Y, Fukuda H, Nishio A, Kawasaki Y, Kuroda A, Suzuki K, Kamidani R, Okamoto H, Fukuta T, Kitagawa Y, Miyake T, Nakane K, Suzuki A, Yoshida T, Tetsuka N, Yoshida S, Koie T, Ogura S. Recombinant antithrombin attenuates acute kidney injury associated with rhabdomyolysis: an in vivo animal study. Intensive Care Med Exp 2024; 12:7. [PMID: 38282162 PMCID: PMC10822833 DOI: 10.1186/s40635-024-00594-y] [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: 08/24/2023] [Accepted: 01/04/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Rhabdomyolysis is characterized by the destruction and necrosis of skeletal muscle tissue, resulting in acute kidney injury (AKI). Recombinant antithrombin (rAT) has DNA repair and vascular endothelial-protection properties. Herein, we investigated whether rAT therapy has beneficial effects against rhabdomyolysis-induced AKI. Ten-week-old male B6 mice were injected with 5 mL/kg of 50% glycerol intramuscularly in the left thigh after 24 h of fasting to create a rhabdomyolysis mouse model. Further, 750 IU/kg rAT was injected intraperitoneally at 24 and 72 h after the rhabdomyolysis model was established. The mice were euthanized after 96 h for histological analysis. Saline was administered to mice in the control group. RESULTS Blood tests show elevated serum creatinine, urea nitrogen, and neutrophil gelatinase-associated lipocalin levels in rhabdomyolysis. Loss of tubular epithelial cell nuclei and destruction of the tubular luminal surface structure was observed in the untreated group, which improved with rAT treatment. Immunostaining for Ki-67 showed increased Ki-67-positive nuclei in the tubular epithelial cells in the rAT group, suggesting that rAT may promote tubular epithelial cell regeneration. The microvilli of the brush border of the renal tubules were shed during rhabdomyolysis, and rAT treatment reduced this injury. The vascular endothelial glycocalyx, which is usually impaired by rhabdomyolysis, became functional following rAT treatment. CONCLUSIONS Treatment with rAT suppressed rhabdomyolysis-induced AKI, suggesting that rAT therapy may be a novel therapeutic approach.
Collapse
Affiliation(s)
- Tomotaka Miura
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Tomoki Okuda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Kodai Suzuki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
- Department of Infection Control, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan.
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan.
| | - Hiroyuki Tomita
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan.
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Chihiro Takada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Kosuke Mori
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Hirotaka Asano
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Soichiro Kano
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Yugo Wakayama
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Yohei Fukuda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Hirotsugu Fukuda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Ayane Nishio
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Yuki Kawasaki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Ayumi Kuroda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Keiko Suzuki
- Department of Infection Control, Gifu University Graduate School of Medicine, Gifu, Japan
- Department of Pharmacy, Gifu University Hospital, Gifu, Japan
| | - Ryo Kamidani
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Haruka Okamoto
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
- Abuse Prevention Center, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tetsuya Fukuta
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Yuichiro Kitagawa
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Takahito Miyake
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Keita Nakane
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akio Suzuki
- Department of Pharmacy, Gifu University Hospital, Gifu, Japan
| | - Takahiro Yoshida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Nobuyuki Tetsuka
- Department of Infection Control, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shozo Yoshida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
- Abuse Prevention Center, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takuya Koie
- Department of Urology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shinji Ogura
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
5
|
Chung FT, Kuo CH, Wang CH, Lin SM. Thrombin worsens extravascular lung water and outcomes of septic patients with acute respiratory distress syndrome: A case control study. Medicine (Baltimore) 2023; 102:e36200. [PMID: 38050307 PMCID: PMC10695594 DOI: 10.1097/md.0000000000036200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/27/2023] [Indexed: 12/06/2023] Open
Abstract
Endothelial cell (EC) activation may increase systemic vascular permeability, causing extravascular lung water (EVLW) in sepsis with acute respiratory distress syndrome (ARDS). However, the correlation between thrombin and EVLW in sepsis and ARDS has not yet been addressed. Patients with sepsis and ARDS were prospectively enrolled between 2014 and 2016, and EVLW and serum thrombin levels on days 1 and 3 were measured and compared between surviving and non-surviving patients. Additionally, morphological changes in human umbilical vein endothelial cells (HUVECs) in the serum of patients with high and low EVLW were evaluated. The levels of EVLW, endothelial cells, and thrombin may positively correlate with the survival of patients with severe sepsis and ARDS. Twenty-seven patients were enrolled, and baseline characteristics, including age, sex, Acute Physiology and Chronic Health Evaluation (APACHE) II, prior 24-h fluid balance, body mass index, and shock status, were similar between survivors and non-survivors; however, day 1 EVLW was higher in non-survivors (27.5 ± 8.4 vs 22 ± 6.5 mL/kg, P = .047). EVLW of survivors improved from day 1 to day 3 (22 ± 6.5 vs 11 ± 3.8 mL/kg, P < .001), but did not improve in non-survivors (27.5 ± 8.4 vs 28 ± 6.7 mL/kg, P = .086), which means that patients had significantly lower EVLW on day 3 than on day 1. Thrombin levels of survivors significantly improved (1.03 ± 0.55 vs 0.87 ± 0.25 U/mL, P = .04) but did not improve in non-survivors (1.97 ± 0.75 vs 2.2 ± 0.75 U/mL, P = .08) from day 1 to day 3. EVLW and thrombin levels were positively correlated (r2 = 0.71, P < .0001). In vitro, the morphology and junctions of HUVECs changed when the serum from patients with high EVLW was added. The intercellular distances among the control, high EVLW, and low EVLW groups were 5.25 ± 1.22, 21.33 ± 2.15, and 11.17 ± 1.64 µm, respectively (P < .05).
Collapse
Affiliation(s)
- Fu-Tsai Chung
- Department of Thoracic Medicine, New Taipei Municipal TuCheng Hospital, New Taipei, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University, College of Medicine, Taipei, Taiwan
- Department of Respiratory Therapy, New Taipei Municipal TuCheng Hospital, New Taipei, Taiwan
- Scholar of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Thoracic Medicine, St’ Paul Hospital, Taoyuan, Taiwan
| | - Chih-Hsi Kuo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University, College of Medicine, Taipei, Taiwan
| | - Chun-Hua Wang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University, College of Medicine, Taipei, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University, College of Medicine, Taipei, Taiwan
| |
Collapse
|
6
|
Li T, Bo F, Meng X, Wang D, Ma J, Dai Z. The effect of perioperative antithrombin supplementation on blood conservation and postoperative complications after cardiopulmonary bypass surgery: A systematic review, meta-analysis and trial sequential analysis. Heliyon 2023; 9:e22266. [PMID: 38053853 PMCID: PMC10694320 DOI: 10.1016/j.heliyon.2023.e22266] [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: 04/07/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023] Open
Abstract
Study objective Antithrombin (AT) activity is reduced during cardiopulmonary bypass (CPB) surgery. Guidelines has demonstrated that perioperative AT supplementation contributed to blood conservation and prevent perioperative thrombotic complications and target organ injury owing to its role in reducing thrombin generation. But these recommends is lack of support of meta-analysis in the guidelines. This meta-analysis aims to include all the relevant randomized controlled trails (RCT) on patients who experienced cardiac surgeries with CPB and investigate the effect of perioperative AT on blood conservation and complications after cardiac surgery. Methods Standard published RCTs were searched from bibliographic databases to identify all evidence reporting perioperative AT supplementation for patients undergoing cardiovascular surgeries. The primary outcome was postoperative blood loss, the secondary outcomes were blood component transfusion (red blood cell (RBC), fresh frozen plasma (FFP), platelet and autologous blood), postoperative morbidity and in hospital mortality. The relative risk (RR) for dichotomous outcomes and the standardized mean difference (SMD) for continuous outcomes were estimated using a random-effects model. Trial sequential analysis (TSA) was performed using TSA software 0.9.5.10. Results 13 RCTs with 996 participants undergoing different cardiovascular surgeries were included. Meta-analysis showed AT did not decrease postoperative blood loss (SMD -0.01, 95%CI -0.2 to 0.19). Subgroup analysis showed the effect of AT on postoperative blood loss was not associated with age, RCT type, surgery type, injection time of AT and AT deficiency. TSA further suggested that no additional studies were required for the stable result. Perioperative AT also did not reduce RBC ((SMD 0.10, 95%CI -0.66 to 0.85), (RR 0.99, 95%CI 0.83 to 1.19)), FFP ((SMD 0.11, 95%CI -0.19 to 0.41), (RR 1.30, 95%CI 0.90 to 1.87)), platelet (RR 1.10, 95%CI 0.83 to 1.46) and autologous blood (SMD 0.46, 95%CI -0.12 to 1.8504) transfusions. Perioperative AT significantly increased in hospital mortality (RR 2.53, 95%CI 1.02 to 6.28) and acute kidney injury (AKI) (RR 3.72, 95%CI 1.73 to 8.04) incidence. There was no significant difference in postoperative reexploration, thromboembolism, ECMO/IABP support, and stroke incidence between AT and non-AT group. Conclusions With the improvement of AT level and heparin sensitivity, perioperative AT has no significant effect on blood conservation. And it is noteworthy that the treatment increased in hospital mortality and the incidence of AKI after cardiac surgery.
Collapse
Affiliation(s)
- Tao Li
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - FengShan Bo
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - XiangRui Meng
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Di Wang
- Department of Internal Medicine, Yantai Haigang Hospital, Yantai, 264000, Shandong, China
| | - Jiahai Ma
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Zhao Dai
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| |
Collapse
|
7
|
Wang Y, Schneider SW, Gorzelanny C. Crosstalk between Circulating Tumor Cells and Plasma Proteins-Impact on Coagulation and Anticoagulation. Cancers (Basel) 2023; 15:cancers15113025. [PMID: 37296987 DOI: 10.3390/cancers15113025] [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: 04/04/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Cancer metastasis is a complex process. After their intravasation into the circulation, the cancer cells are exposed to a harsh environment of physical and biochemical hazards. Whether circulating tumor cells (CTCs) survive and escape from blood flow defines their ability to metastasize. CTCs sense their environment with surface-exposed receptors. The recognition of corresponding ligands, e.g., fibrinogen, by integrins can induce intracellular signaling processes driving CTCs' survival. Other receptors, such as tissue factor (TF), enable CTCs to induce coagulation. Cancer-associated thrombosis (CAT) is adversely connected to patients' outcome. However, cancer cells have also the ability to inhibit coagulation, e.g., through expressing thrombomodulin (TM) or heparan sulfate (HS), an activator of antithrombin (AT). To that extent, individual CTCs can interact with plasma proteins, and whether these interactions are connected to metastasis or clinical symptoms such as CAT is largely unknown. In the present review, we discuss the biological and clinical relevance of cancer-cell-expressed surface molecules and their interaction with plasma proteins. We aim to encourage future research to expand our knowledge of the CTC interactome, as this may not only yield new molecular markers improving liquid-biopsy-based diagnostics but also additional targets for better cancer therapies.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Christian Gorzelanny
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
8
|
Moront MG, Woodward MK, Essandoh MK, Avery EG, Reece TB, Brzezinski M, Spiess B, Shore-Lesserson L, Chen J, Henriquez W, Barceló M, Despotis G, Karkouti K, Levy JH, Ranucci M, Mondou E. A Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial of Preoperative Antithrombin Supplementation in Patients at Risk for Antithrombin Deficiency After Cardiac Surgery. Anesth Analg 2022; 135:757-768. [PMID: 35877927 DOI: 10.1213/ane.0000000000006145] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Antithrombin (AT) activity is reduced during cardiac operations with cardiopulmonary bypass (CPB), which is associated with adverse outcomes. Preoperative AT supplementation, to achieve >58% and <100% AT activity, may potentially reduce postoperative morbidity and mortality in cardiac operations with CPB. This prospective, multicenter, randomized, double-blind, placebo-controlled study was designed to evaluate the safety and efficacy of preoperative treatment with AT supplementation in patients at risk for low AT activity after undergoing cardiac surgery with CPB. METHODS A total of 425 adult patients were randomized (1:1) to receive either a single dose of AT (n = 213) to achieve an absolute increase of 20% above pretreatment AT activity or placebo (n = 212) before surgery. The study duration was approximately 7 weeks. The primary efficacy end point was the percentage of patients with any component of a major morbidity composite (postoperative mortality, stroke, acute kidney injury [AKI], surgical reexploration, arterial or venous thromboembolic events, prolonged mechanical ventilation, and infection) in the 2 groups. Secondary end points included AT activity, blood loss, transfusion requirements, duration of intensive care unit (ICU), and hospital stays. Safety was also assessed. RESULTS Overall, 399 patients (men, n = 300, 75.2%) with a mean (standard deviation [SD]) age of 66.1 (11.7) years, with the majority undergoing complex surgical procedures (n = 266, 67.9%), were analyzed. No differences in the percentage of patients experiencing morbidity composite outcomes between groups were observed (AT-treated 68/198 [34.3%] versus placebo 58/194 [29.9%]; P = .332; relative risk, 1.15). After AT infusion, AT activity was significantly higher in the AT group (108% [42-143]) versus placebo group (76% [40-110]), and lasted up to postoperative day 2. At ICU, the frequency of patients with AT activity ≥58% in the AT group (81.5%) was significantly higher (P < .001) versus placebo group (43.2%). Secondary end point analysis did not show any advantage of AT over placebo group. There were significantly more patients with AKI (P < .001) in the AT group (23/198; 11.6%) than in the placebo group (5/194, 2.6%). Safety results showed no differences in treatment-emergent adverse events nor bleeding events between groups. CONCLUSIONS AT supplementation did not attenuate adverse postoperative outcomes in our cohort of patients undergoing cardiac surgery with CPB.
Collapse
Affiliation(s)
- Michael George Moront
- From the, Department of Cardiothoracic Sugery, Promedical Toledo Hospital, Toledo, Ohio
| | | | - Michael K Essandoh
- Department of Anesthesiology' Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Edwin G Avery
- Department of Anesthesiology and Perioperative Medicine, University Hospital Case Medical Center, Cleveland, Ohio
| | - T Brett Reece
- Department of Surgery' Division of Cardiothoracic Surgery, University of Colorado, Aurora, Colorado
| | - Marek Brzezinski
- Department of Anesthesiology and Perioperative Care, University of California, San Francisco, California.,San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Bruce Spiess
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, Florida
| | | | - Junliang Chen
- Bioscience Research Group, Grifols, Barcelona, Spain
| | | | | | - George Despotis
- Departments of Pathology, Immunology and Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Keyvan Karkouti
- Department of Anesthesia and Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jerrold H Levy
- Department of Anesthesiology and Critical Care, Duke University School of Medicine, Durham, North Carolina
| | - Marco Ranucci
- Department of Cardiothoracic and Vascular Anesthesia and Intensive Care, IRCSS Policlinico San Donato, Milan, Italy
| | - Elsa Mondou
- Bioscience Research Group, Grifols, Barcelona, Spain
| | | | | |
Collapse
|
9
|
Razanakolona M, Adam F, Bianchini E, Saller F, Carvalho AD, Diehl JL, Denis CV, Meziani F, Borgel D, Helms J, Vasse M. Anti-inflammatory Activity of the Protein Z-Dependent Protease Inhibitor. TH OPEN 2021; 5:e220-e229. [PMID: 34189397 PMCID: PMC8233056 DOI: 10.1055/s-0041-1730037] [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: 09/28/2020] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
The protein Z (PZ)-dependent plasma protease inhibitor (ZPI) is a glycoprotein that inhibits factor XIa and, in the presence of PZ, FXa. Recently, ZPI has been shown to be an acute-phase protein (APP). As usually APPs downregulate the harmful effects of inflammation, we tested whether ZPI could modulate the increase of cytokines observed in inflammatory states. We observed that recombinant human ZPI (rhZPI) significantly decreases the levels of interleukin (IL)-1, IL-6, and tumor necrosis factor- α (TNF-α) induced by lipopolysaccharide (LPS) in a whole blood model. This inhibitory effect was unaffected by the presence of PZ or heparin. A ZPI mutant within the reactive loop center ZPI (Y387A), lacking anticoagulant activity, still had an anti-inflammatory activity. Surprisingly, rhZPI did not inhibit the synthesis of IL-6 or TNF-α when purified monocytes were stimulated by LPS, whereas the inhibitory effect was evidenced when lymphocytes were added to monocytes. The requirement of lymphocytes could be due to the synthesis of CCL5 (RANTES), a chemokine mainly produced by activated lymphocytes which is induced by rhZPI, and which can reduce the production of proinflammatory cytokines in whole blood. Lastly, we observed that the intraperitoneal injection of rhZPI significantly decreased LPS-induced IL-6 and TNF-α production in mouse plasma.
Collapse
Affiliation(s)
- Mahita Razanakolona
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Frédéric Adam
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Elsa Bianchini
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - François Saller
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Allan de Carvalho
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Jean-Luc Diehl
- Département de réanimation médicale, Hôpital Européen Georges Pompidou, Paris, France
| | - Cécile V Denis
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Ferhat Meziani
- Faculté de Médecine, Service de Médecine Intensive-Réanimation, Université de Strasbourg (UNISTRA), Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Strasbourg, France.,INSERM (French National Institute of Health and Medical Research), Regenerative Nanomedicine (RNM), FMTS, Strasbourg, France
| | - Delphine Borgel
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France.,APHP, Laboratoire d'Hématologie, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Julie Helms
- Faculté de Médecine, Service de Médecine Intensive-Réanimation, Université de Strasbourg (UNISTRA), Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Strasbourg, France.,ImmunoRhumatologie Moléculaire, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Marc Vasse
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France.,Service de Biologie Clinique, Hôpital Foch, Suresnes, France
| |
Collapse
|
10
|
Fang XZ, Wang YX, Xu JQ, He YJ, Peng ZK, Shang Y. Immunothrombosis in Acute Respiratory Dysfunction of COVID-19. Front Immunol 2021; 12:651545. [PMID: 34149692 PMCID: PMC8207198 DOI: 10.3389/fimmu.2021.651545] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/12/2021] [Indexed: 01/10/2023] Open
Abstract
COVID-19 is an acute, complex disorder that was caused by a new β-coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Based on current reports, it was surprising that the characteristics of many patients with COVID-19, who fulfil the Berlin criteria for acute respiratory distress syndrome (ARDS), are not always like those of patients with typical ARDS and can change over time. While the mechanisms of COVID-19–related respiratory dysfunction in COVID-19 have not yet been fully elucidated, pulmonary microvascular thrombosis is speculated to be involved. Considering that thrombosis is highly related to other inflammatory lung diseases, immunothrombosis, a two-way process that links coagulation and inflammation, seems to be involved in the pathophysiology of COVID-19, including respiratory dysfunction. Thus, the current manuscript will describe the proinflammatory milieu in COVID-19, summarize current evidence of thrombosis in COVID-19, and discuss possible interactions between these two.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Qain Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Jun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe-Kang Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Peñas-Martínez J, Luengo-Gil G, Espín S, Bohdan N, Ortega-Sabater C, Ródenas MC, Zaragoza-Huesca D, López-Andreo MJ, Plasencia C, Vicente V, Carmona-Bayonas A, Martínez-Martínez I. Anti-Tumor Functions of Prelatent Antithrombin on Glioblastoma Multiforme Cells. Biomedicines 2021; 9:biomedicines9050523. [PMID: 34067120 PMCID: PMC8151964 DOI: 10.3390/biomedicines9050523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/27/2021] [Accepted: 05/04/2021] [Indexed: 11/24/2022] Open
Abstract
Antithrombin, the main physiological inhibitor of the coagulation cascade, exerts anti-tumor effects on glioblastoma multiforme cells. Antithrombin has different conformations: native, heparin-activated, prelatent, latent, and cleaved. The prelatent form has an intermediate affinity between latent and native antithrombin, although it is the most antiangiogenic form. Herein, we investigate the effect of this conformation on the tumorigenic processes of glioblastoma multiforme cells. Antithrombin forms were purified by chromatography. Chromogenic/fluorogenic assays were carried out to evaluate enteropeptidase and hepsin inhibition, two serine proteases involved in these processes. Wound healing, Matrigel invasion and BrdU incorporation assays were performed to study migration, invasion and proliferation. E-cadherin, Vimentin, VEGFA, pAKT, STAT3, pSTAT3, and pERK1/2 expression was assessed by Western blot and/or qRT-PCR. Prelatent antithrombin inhibited both enteropeptidase and hepsin, although it was less efficient than the native conformation. Exposure to prelatent antithrombin significantly reduced migration and invasion but not proliferation of U-87 MG, being the conformation most efficient on migration. Prelatent antithrombin down-regulated VEGFA, pSTAT3, and pERK1/2 expression in U-87 MG cells. Our work elucidates that prelatent antithrombin has surprisingly versatile anti-tumor properties in U-87 MG glioblastoma multiforme cells. This associates with resistance pathway activation, the decreased expression of tumorigenic proteins, and increased angiogenesis, postulating the existence of a new, formerly unknown receptor with potential therapeutic implications.
Collapse
Affiliation(s)
- Julia Peñas-Martínez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
| | - Ginés Luengo-Gil
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
| | - Salvador Espín
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
| | - Nataliya Bohdan
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
| | - Carmen Ortega-Sabater
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
| | - Maria Carmen Ródenas
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
| | - David Zaragoza-Huesca
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
| | - María José López-Andreo
- Sección de Biología Molecular, El Área Científica y Técnica de Investigación (ACTI), Universidad de Murcia, 30003 Murcia, Spain;
| | - Carme Plasencia
- Applied Research Using Omic Sciences S.L., 08028 Barcelona, Spain;
| | - Vicente Vicente
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, U-765-CIBERER, Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Alberto Carmona-Bayonas
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
- Correspondence: (A.C.-B.); (I.M.-M.); Tel.: +34-9683-41990 (A.C.-B. & I.M.-M.)
| | - Irene Martínez-Martínez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, 30003 Murcia, Spain; (J.P.-M.); (G.L.-G.); (S.E.); (N.B.); (C.O.-S.); (M.C.R.); (D.Z.-H.); (V.V.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, U-765-CIBERER, Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Correspondence: (A.C.-B.); (I.M.-M.); Tel.: +34-9683-41990 (A.C.-B. & I.M.-M.)
| |
Collapse
|
12
|
Antithrombin and Its Role in Host Defense and Inflammation. Int J Mol Sci 2021; 22:ijms22084283. [PMID: 33924175 PMCID: PMC8074369 DOI: 10.3390/ijms22084283] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Antithrombin (AT) is a natural anticoagulant that interacts with activated proteases of the coagulation system and with heparan sulfate proteoglycans (HSPG) on the surface of cells. The protein, which is synthesized in the liver, is also essential to confer the effects of therapeutic heparin. However, AT levels drop in systemic inflammatory diseases. The reason for this decline is consumption by the coagulation system but also by immunological processes. Aside from the primarily known anticoagulant effects, AT elicits distinct anti-inflammatory signaling responses. It binds to structures of the glycocalyx (syndecan-4) and further modulates the inflammatory response of endothelial cells and leukocytes by interacting with surface receptors. Additionally, AT exerts direct antimicrobial effects: depending on AT glycosylation it can bind to and perforate bacterial cell walls. Peptide fragments derived from proteolytic degradation of AT exert antibacterial properties. Despite these promising characteristics, therapeutic supplementation in inflammatory conditions has not proven to be effective in randomized control trials. Nevertheless, new insights provided by subgroup analyses and retrospective trials suggest that a recommendation be made to identify the patient population that would benefit most from AT substitution. Recent experiment findings place the role of various AT isoforms in the spotlight. This review provides an overview of new insights into a supposedly well-known molecule.
Collapse
|
13
|
Lavranou GA, Mentzelopoulos S, Katsaounou P, Siempos I, Kalomenidis I, Geranaki A, Routsi C, Zakynthinos S. Can Coagulation System Disorders and Cytokine and Inflammatory Marker Levels Predict the Temporary Clinical Deterioration or Improvement of Septic Patients on ICU Admission? J Clin Med 2021; 10:jcm10081548. [PMID: 33917002 PMCID: PMC8067680 DOI: 10.3390/jcm10081548] [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: 02/01/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 11/16/2022] Open
Abstract
Although coagulation disorders and immune/inflammatory response have been associated with the final outcome of patients with sepsis, their link with thetemporaryclinical deterioration or improvement of patients is unknown. We aimed to investigate this link. We prospectively included consecutive patients admitted to the intensive care unit (ICU) with a suspected diagnosis of infection and evaluated within the first 24 h from admission. Blood levels of many cytokines and inflammatory and coagulation factors were measured and their predictive value was assessed by calculating the Area Under the Receiver Operating Characteristic (AUROC) curves. Patients (n = 102) were allocated in five groups, i.e., sepsis (n = 14), severe sepsis (n = 17), septic shock (n = 28), Systemic Inflammatory Response Syndrome (SIRS) without infection (n = 17), and trauma/surgery without SIRS or infection (n = 26). In septic shock, coagulation factors FVII and FIX and Protein C had AUROCs 0.67-0.78. In severe sepsis, Antithrombin III, Protein C, C-reactive protein, Procalcitonin and Thrombopoietin had AUROCs 0.73-0.75. In sepsis, Tumor Necrosis Factor a, and Interleukins 1β and 10 had AUROCs 0.66-0.72. In patients admitted to the ICU with a suspected diagnosis of infection, coagulation factors and inhibitors, as well as cytokine and inflammatory marker levels, have substantial predictive value in distinct groups of septic patients.
Collapse
Affiliation(s)
- Georgia-Athanasia Lavranou
- First Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, ‘Evangelismos’ Hospital, 45-47 Ipsilandou St, GR-10675 Athens, Greece; (G.-A.L.); (S.M.); (P.K.); (I.S.); (I.K.); (C.R.)
| | - Spyros Mentzelopoulos
- First Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, ‘Evangelismos’ Hospital, 45-47 Ipsilandou St, GR-10675 Athens, Greece; (G.-A.L.); (S.M.); (P.K.); (I.S.); (I.K.); (C.R.)
| | - Paraskevi Katsaounou
- First Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, ‘Evangelismos’ Hospital, 45-47 Ipsilandou St, GR-10675 Athens, Greece; (G.-A.L.); (S.M.); (P.K.); (I.S.); (I.K.); (C.R.)
| | - Ilias Siempos
- First Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, ‘Evangelismos’ Hospital, 45-47 Ipsilandou St, GR-10675 Athens, Greece; (G.-A.L.); (S.M.); (P.K.); (I.S.); (I.K.); (C.R.)
| | - Ioannis Kalomenidis
- First Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, ‘Evangelismos’ Hospital, 45-47 Ipsilandou St, GR-10675 Athens, Greece; (G.-A.L.); (S.M.); (P.K.); (I.S.); (I.K.); (C.R.)
| | - Aikaterini Geranaki
- Hematology Laboratory, ‘Evangelismos’ Hospital, 45-47 Ipsilandou St, GR-10675 Athens, Greece;
| | - Christina Routsi
- First Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, ‘Evangelismos’ Hospital, 45-47 Ipsilandou St, GR-10675 Athens, Greece; (G.-A.L.); (S.M.); (P.K.); (I.S.); (I.K.); (C.R.)
| | - Spyros Zakynthinos
- First Department of Intensive Care Medicine, School of Medicine, National and Kapodistrian University of Athens, ‘Evangelismos’ Hospital, 45-47 Ipsilandou St, GR-10675 Athens, Greece; (G.-A.L.); (S.M.); (P.K.); (I.S.); (I.K.); (C.R.)
- Correspondence:
| |
Collapse
|
14
|
Antithrombin III Contributes to the Protective Effects of Fresh Frozen Plasma Following Hemorrhagic Shock by Preventing Syndecan-1 Shedding and Endothelial Barrier Disruption. Shock 2021; 53:156-163. [PMID: 31389906 DOI: 10.1097/shk.0000000000001432] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Endothelial dysfunction during hemorrhagic shock (HS) is associated with loss of cell-associated syndecan-1 (Sdc1) and hyperpermeability. Fresh frozen plasma (FFP) preserves Sdc1 and reduces permeability following HS, although the key mediators remain unknown. Antithrombin III (ATIII) is a plasma protein with potent anti-inflammatory and endothelial protective activity. We hypothesized that the protective effects of FFP on endothelial Sdc1 and permeability are mediated, in part, through ATIII. METHODS ATIII and Sdc1 were measured in severely injured patients upon admission (N = 125) and hospital day 3 (N = 90) for correlation analysis. In vitro effects of ATIII on human lung microvascular endothelial cells (HLMVECs) were determined by pretreating cells with vehicle, FFP, ATIII-deficient FFP, or purified ATIII followed by TNFα stimulation. Sdc1 expression was measured by immunostaining and permeability by electrical impedance. To determine the role of ATIII in vivo, male mice were subjected to a fixed pressure exsanguination model of HS, followed by resuscitation with FFP, ATIII-deficient FFP, or ATIII-deficient FFP with ATIII repletion. Lung Sdc1 expression was assessed by immunostaining. RESULTS Pearson correlation analysis showed a significant negative correlation between plasma levels of Sdc1 and ATIII (R = -0.62; P < 0.0001) in injured patients on hospital day 3. Also, in vitro, FFP and ATIII prevented TNFα-induced permeability (P < 0.05 vs TNFα) in HLMVECs. ATIII-deficient FFP had no effect; however, ATIII restoration reestablished its protective effects in a dose-dependent manner. Similarly, FFP and ATIII prevented TNFα-induced Sdc1 shedding in HLMVECs; however, ATIII-deficient FFP did not. In mice, Sdc1 expression was increased following FFP resuscitation (1.7 ± 0.5, P < 0.01) vs. HS alone (1.0 ± 0.3); however, no improvement was seen following ATIII-deficient FFP treatment (1.3 ± 0.4, P = 0.3). ATIII restoration improved Sdc1 expression (1.5 ± 0.9, P < 0.05) similar to that of FFP resuscitation. CONCLUSIONS ATIII plays a role in FFP-mediated protection of endothelial Sdc1 expression and barrier function, making it a potential therapeutic target to mitigate HS-induced endothelial dysfunction. Further studies are needed to elucidate the mechanisms by which ATIII protects the endothelium.
Collapse
|
15
|
Anaklı İ, Ergin Özcan P, Polat Ö, Orhun G, Alay GH, Tuna V, Çeliksoy E, Kılıç M, Mercan M, Ali A, Beşışık S, Esen F. Prognostic Value of Antithrombin Levels in COVID-19 Patients and Impact of Fresh Frozen Plasma Treatment: A Retrospective Study. Turk J Haematol 2021; 38:15-21. [PMID: 33486940 PMCID: PMC7927448 DOI: 10.4274/tjh.galenos.2021.2020.0695] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective: The defective interplay between coagulation and inflammation may be the leading cause of intravascular coagulation and organ dysfunction in coronavirus disease-19 (COVID-19) patients. Abnormal coagulation profiles were reported to be associated with poor outcomes. In this study, we assessed the prognostic values of antithrombin (AT) activity levels and the impact of fresh frozen plasma (FFP) treatment on outcome. Materials and Methods: Conventional coagulation parameters as well as AT activity levels and outcomes of 104 consecutive critically ill acute respiratory distress syndrome (ARDS) patients with laboratory-confirmed COVID-19 disease were retrospectively analyzed. Patients with AT activity below 75% were treated with FFP. Maximum AT activity levels achieved in those patients were recorded. Results: AT activity levels at admission were significantly lower in nonsurvivors than survivors (73% vs. 81%). The cutoff level for admission AT activity was 79% and 58% was the lowest AT for survival. The outcome in those patients who had AT activity levels above 75% after FFP treatment was better than that of the nonresponding group. As well as AT, admission values of D-dimer, C-reactive protein, and procalcitonin were coagulation and inflammatory parameters among the mortality risk factors. Conclusion: AT activity could be used as a prognostic marker for survival and organ failure in COVID-19-associated ARDS patients. AT supplementation therapy with FFP in patients with COVID-19-induced hypercoagulopathy may improve thrombosis prophylaxis and thus have an impact on survival.
Collapse
Affiliation(s)
- İlkay Anaklı
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Perihan Ergin Özcan
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Özlem Polat
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Günseli Orhun
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Gülçin Hilal Alay
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Verda Tuna
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Emre Çeliksoy
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Mehmet Kılıç
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Mutlu Mercan
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Achmet Ali
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| | - Sevgi Beşışık
- İstanbul University, İstanbul Faculty of Medicine, Department of Internal Medicine, Division of Hematology, İstanbul, Turkey
| | - Figen Esen
- İstanbul University, İstanbul Faculty of Medicine, Department of Anesthesiology and Reanimation, İstanbul, Turkey
| |
Collapse
|
16
|
Dolmatova EV, Wang K, Mandavilli R, Griendling KK. The effects of sepsis on endothelium and clinical implications. Cardiovasc Res 2021; 117:60-73. [PMID: 32215570 PMCID: PMC7810126 DOI: 10.1093/cvr/cvaa070] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/03/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Sepsis accounts for nearly 700 000 deaths in Europe annually and is caused by an overwhelming host response to infection resulting in organ failure. The endothelium is an active contributor to sepsis and as such represents a major target for therapy. During sepsis, endothelial cells amplify the immune response and activate the coagulation system. They are both a target and source of inflammation and serve as a link between local and systemic immune responses. In response to cytokines produced by immune cells, the endothelium expresses adhesion molecules and produces vasoactive compounds, inflammatory cytokines, and chemoattractants, thus switching from an anticoagulant to procoagulant state. These responses contribute to local control of infection, but systemic activation can lead to microvascular thrombosis, capillary permeability, hypotension, tissue hypoxia, and ultimately tissue damage. This review focuses on the role of the endothelium in leucocyte adhesion and transmigration as well as production of reactive oxygen and nitrogen species, microRNAs and cytokines, formation of signalling microparticles, and disseminated intravascular coagulation. We also discuss alterations in endothelial permeability and apoptosis. Finally, we review the diagnostic potential of endothelial markers and endothelial pathways as therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Elena V Dolmatova
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Keke Wang
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Rohan Mandavilli
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| |
Collapse
|
17
|
Rezaie AR, Giri H. Anticoagulant and signaling functions of antithrombin. J Thromb Haemost 2020; 18:3142-3153. [PMID: 32780936 PMCID: PMC7855051 DOI: 10.1111/jth.15052] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/25/2020] [Accepted: 08/04/2020] [Indexed: 12/19/2022]
Abstract
Antithrombin (AT) is a major plasma glycoprotein of the serpin superfamily that regulates the proteolytic activity of the procoagulant proteases of both intrinsic and extrinsic pathways. Two important structural features that participate in the regulatory function of AT include a mobile reactive center loop that binds to active site of coagulation proteases, trapping them in the form of inactive covalent complexes, and a basic D-helix that binds to therapeutic heparins and heparan sulfate proteoglycans (HSPGs) on vascular endothelial cells. The binding of D-helix of AT by therapeutic heparins promotes the reactivity of the serpin with coagulation proteases by several orders of magnitude by both a conformational activation of the serpin and a template (bridging) mechanism. In addition to its essential anticoagulant function, AT elicits a potent anti-inflammatory signaling response when it binds to distinct vascular endothelial cell HSPGs, thereby inducing prostacyclin synthesis. Syndecans-4 has been found as a specific membrane-bound HSPG receptor on endothelial cells that relays the signaling effect of AT to the relevant second messenger molecules in the signal transduction pathways inside the cell. However, following cleavage by coagulation proteases and/or by spontaneous conversion to a latent form, AT loses both its anti-inflammatory activity and high-affinity interaction with heparin and HSPGs. Interestingly, these low-affinity heparin conformers of AT elicit potent proapoptotic and antiangiogenic activities by also binding to specific HSPGs by unknown mechanisms. This review article will summarize current knowledge about mechanisms through which different conformers of AT exert their serine protease inhibitory and intracellular signaling functions in these biological pathways.
Collapse
Affiliation(s)
- Alireza R. Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Hemant Giri
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| |
Collapse
|
18
|
Okimoto S, Tashiro H, Iwako H, Kuroda S, Kobayashi T, Hinoi T, Ohdan H. Antithrombin attenuates the progression of hepatocellular carcinoma by regulating neutrophil/interleukin-8 signaling. Hepatol Res 2020; 50:1284-1296. [PMID: 32893935 DOI: 10.1111/hepr.13558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
AIM Inflammation plays an important role in hepatocellular carcinoma (HCC) progression. Here, we examined whether antithrombin (AT) plays a role in attenuating HCC progression, via its anti-inflammatory effects. METHODS HCCs were developed in AT-insufficient (AT+/- ) mice and wild-type (AT+/+ ) mice treated with diethyl nitrosamine and carbon tetrachloride. AT was administered to AT+/- mice. The development of HCC was compared between the three groups. In vitro study, migration assay was performed. The association of the prognosis of patients with HCC and plasma AT values was clinically examined. RESULTS AT suppressed the release of interleukin (IL)-8 from lipopolysaccharide (LPS)-stimulated human neutrophils in vitro. Huh-7 cells that were co-cultured with neutrophils and stimulated with LPS showed significantly enhanced migration; however, Huh-7 cells co-cultured with LPS/AT-stimulated neutrophils showed significantly decreased migration. Moreover, the addition of anti-IL-8 antibodies to LPS-stimulated Huh-7 cells co-cultured with neutrophils also suppressed migration. AT+/- mice (AT plasma activity: 64%) promoted liver cancer, as compared with wild-type mice (AT plasma activity: 135%); AT administration attenuated liver cancer in AT+/- mice. Patients with HCC with a preoperative AT level of ≥70% showed better outcomes after liver resection, as compared with those with an AT level of <70%. IL-8 expression and neutrophil infiltration in HCC tissues were negatively correlated with the AT level. CONCLUSIONS AT attenuates HCC progression by regulating neutrophil/IL-8 signaling.
Collapse
Affiliation(s)
- Sho Okimoto
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Hirotaka Tashiro
- Department of Surgery, National Hospital Organization Kure Medical Center, Kure City, Hiroshima, Japan
| | - Hiroshi Iwako
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Shintaro Kuroda
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Takao Hinoi
- Department of clinical and molecular Genetics, Hiroshima University Hospital, Kasumi, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| |
Collapse
|
19
|
Panicker SR, Biswas I, Giri H, Cai X, Rezaie AR. PKC (Protein Kinase C)-δ Modulates AT (Antithrombin) Signaling in Vascular Endothelial Cells. Arterioscler Thromb Vasc Biol 2020; 40:1748-1762. [PMID: 32404004 DOI: 10.1161/atvbaha.120.314479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Native and latent conformers of AT (antithrombin) induce anti-inflammatory and proapoptotic signaling activities, respectively, in vascular endothelial cells by unknown mechanisms. Synd-4 (syndecan-4) has been identified as a receptor that is involved in transmitting signaling activities of AT in endothelial cells. Approach and Results: In this study, we used flow cytometry, signaling assays, immunoblotting and confocal immunofluorescence microscopy to investigate the mechanism of the paradoxical signaling activities of high-affinity heparin (native) and low-affinity heparin (latent) conformers of AT in endothelial cells. We discovered that native AT binds to glycosaminoglycans on vascular endothelial cells via its heparin-binding D-helix to induce anti-inflammatory signaling responses by recruiting PKC (protein kinase C)-δ to the plasma membrane and promoting phosphorylation of the Synd-4 cytoplasmic domain at Ser179. By contrast, the binding of latent AT to endothelial cells to a site(s), which is not competed by the native AT, induces a proapoptotic effect by localizing PKC-δ to the perinuclear/nuclear compartment in endothelial cells. Overexpression of a dominant-negative form of PKC-δ resulted in inhibition of anti-inflammatory and proapoptotic signaling activities of both native and latent AT. CONCLUSIONS These results indicate that the native and latent conformers of AT may exert their distinct intracellular signaling effects through differentially modulating the subcellular localization of PKC-δ in endothelial cells.
Collapse
Affiliation(s)
- Sumith R Panicker
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (S.R.P., I.B., H.G., X.C., A.R.R.)
| | - Indranil Biswas
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (S.R.P., I.B., H.G., X.C., A.R.R.)
| | - Hemant Giri
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (S.R.P., I.B., H.G., X.C., A.R.R.)
| | - Xiaofeng Cai
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (S.R.P., I.B., H.G., X.C., A.R.R.)
| | - Alireza R Rezaie
- From the Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (S.R.P., I.B., H.G., X.C., A.R.R.).,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City (A.R.R.)
| |
Collapse
|
20
|
Rezaie AR, Giri H. Antithrombin: An anticoagulant, anti-inflammatory and antibacterial serpin. J Thromb Haemost 2020; 18:528-533. [PMID: 32112532 PMCID: PMC7080319 DOI: 10.1111/jth.14724] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Alireza R. Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Hemant Giri
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| |
Collapse
|
21
|
Rehman SU, Ashraf S, Ahamad S, Sarwar T, Husain MA, Ahmad P, Tabish M, Jairajpuri MA. Identification of a novel alternatively spliced isoform of antithrombin containing an additional RCL-like loop. Biochem Biophys Res Commun 2019; 517:421-426. [PMID: 31378371 DOI: 10.1016/j.bbrc.2019.07.113] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/28/2019] [Indexed: 11/22/2022]
Abstract
Antithrombin (AT3) is one of the most important inhibitors of blood coagulation proteases that belong to the serpin family of protease inhibitors. In this study, a novel alternatively spliced isoform of AT3 was identified, both at transcript and protein level. This novel transcript contains an additional region in the continuation of exon 3b that was included in the transcript due to use of an alternate 5' splice site. The existence of the novel transcript was confirmed in human brain and liver through RT-PCR. An analysis of the complete transcript indicated that the native reactive centre loop (RCL) of AT3 is maintained; however the novel amino acid sequence projects out as an additional loop as evident from MD simulation studies. A unique amino acid sequence present in the novel isoform was used for the development of polyclonal antibody. The expression of novel isoform was confirmed in human brain and liver tissue using Western blot analysis. Interestingly an alignment of RCL like domain with other inhibitory serpins showed significant similarity with the neuroserpin RCL. To the best of our knowledge, this is the first evidence of alternatively spliced AT3 sequence containing an additional loop and could have physiological relevance.
Collapse
Affiliation(s)
- Sayeed Ur Rehman
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India; Department of Biochemistry, SCLS, Jamia Hamdard, New Delhi, India
| | - Shazia Ashraf
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India; Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shahzaib Ahamad
- Department of Biotechnology, School of Engineering & Technology, IFTM University, Lodhipur-Rajput, Delhi Road, Moradabad, India
| | - Tarique Sarwar
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | | | - Parvez Ahmad
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Tabish
- Department of Biochemistry, Faculty of Life Sciences, A.M.U, Aligarh, U.P, India
| | | |
Collapse
|
22
|
Juschten J, Ingelse SA, Maas MAW, Girbes ARJ, Juffermans NP, Schultz MJ, Tuinman PR. Antithrombin plus alpha-1 protease inhibitor does not affect coagulation and inflammation in two murine models of acute lung injury. Intensive Care Med Exp 2019; 7:36. [PMID: 31346884 PMCID: PMC6658634 DOI: 10.1186/s40635-019-0240-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 01/23/2023] Open
Abstract
Background In acute respiratory distress syndrome (ARDS), uncontrolled production of activators of coagulation and proinflammatory mediators results in a shift from an adequate local innate immune response to hypercoagulability and inflammation. This study aimed to investigate whether the protease inhibitors antithrombin (AT) and alpha-1 protease inhibitor (A1PI) may attenuate an exaggerated pulmonary immune response. Methods Lung injury was induced either by single intranasal administration of lipopolysaccharide (LPS) (5 mg/kg) in BALB/c mice or by combination of an intravenous injection of LPS (10 mg/kg) with subsequent injurious ventilation using high tidal volumes (12–15 ml/kg) for 4 h in RccHan Wistar rats. Animals received either a single bolus of AT (250 IU/kg) or A1PI (60 mg/kg) alone or in combination, with or without intravenous low-dose heparin (100 U/kg). Control animals received saline. Additional controls received neither LPS, nor ventilation, nor treatment. Endpoints were local and systemic markers of coagulation, e.g., thrombin–antithrombin complexes (TATc), and inflammation, e.g., interleukin-6. Results Both lung injury models resulted in a pronounced immune response within the pulmonary compartment shown by elevated levels of markers of coagulation and inflammation. The two-hit lung injury model also induced profound systemic coagulopathy and inflammation. Monotherapy with AT or A1PI did not reduce pulmonary coagulopathy or inflammation in any lung injury model. Nor did combination therapy with AT and A1PI result in a decrease of coagulation or inflammatory parameters. AT markedly reduced systemic levels of TATc in the two-hit lung injury model. Systemic inflammation was not affected by the different interventions. Additional administration of heparin did not lead to macroscopic bleeding incidences. Conclusions In two different murine models of acute lung injury, neither single therapy with AT or A1PI nor combination of both agents attenuates the pronounced pulmonary coagulation or inflammatory response.
Collapse
Affiliation(s)
- Jenny Juschten
- Department of Intensive Care, Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands. .,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands. .,Department of Intensive Care, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands. .,Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands.
| | - Sarah Anne Ingelse
- Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands.,Emma Children's Hospital-Pediatric Intensive Care Unit, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands
| | - Martinus Adrianus Wilhelmus Maas
- Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands
| | - Armand Roelof Johan Girbes
- Department of Intensive Care, Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands
| | - Nicole Petra Juffermans
- Department of Intensive Care, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands
| | - Marcus Josephus Schultz
- Department of Intensive Care, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands.,Mahidol Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand
| | - Pieter Roel Tuinman
- Department of Intensive Care, Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands
| |
Collapse
|
23
|
Bachler M, Hell T, Schausberger L, Schlömmer C, Schäfer V, Liebensteiner M, Schäffler K, Schenk B, Fries D, Innerhofer P, Niederwanger C. Response patterns of routinely measured inflammatory and coagulatory parameters in sepsis. PeerJ 2019; 7:e7147. [PMID: 31275752 PMCID: PMC6590445 DOI: 10.7717/peerj.7147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 05/19/2019] [Indexed: 12/13/2022] Open
Abstract
Background Sepsis is characterized by a pro-inflammatory and pro-coagulatory shift which can induce life-threatening complications. Close monitoring and risk stratification of sepsis patients is crucial for proper treatment and consequently patient outcome. Therefore, this study focuses on the response patterns of inflammatory and coagulatory parameters used in clinical routines to estimate the course of sepsis. Methods A total of 1,110 patients diagnosed with sepsis were retrospectively analyzed to identify response patterns for risk stratification of routine parameters measured at the peak level of C-reactive protein. Cluster analysis was used and the differences in the patient characteristics and 28-day survival were assessed. Cox proportional hazards regression model for survival stratified by the clusters was performed. Results The analyses revealed the parameters to have five distinct response patterns. These clusters reflect the etiology as well as the course of sepsis associated with different mortalities. Here, impairment of the liver plays a crucial role in the ability to appropriately respond to sepsis. Of the routinely measured parameters, C-reactive protein and antithrombin seem to be unspecific for stratification of septic patients. Adjusted for the individual clusters, survival was associated with an increase in fibrinogen (p = 0.0042), platelets (p = 0.0003) and PT (p = 0.001) as well as a decrease in leukocytes (p = 0.034). Conclusions This study reveals that patients have distinct response patterns of inflammatory and coagulatory parameters depending on disease etiology. These patterns are associated with different mortalities although the patients have similar levels of C-reactive protein. Independently of the type of response, good coagulatory capacity seems to be crucial for patient survival.
Collapse
Affiliation(s)
- Mirjam Bachler
- University for Health Sciences, Medical Informatics and Technology, Institute for Sports Medicine, Alpine Medicine and Health Tourism, Hall, Austria
| | - Tobias Hell
- Department of Mathematics, Faculty of Mathematics, Computer Science and Physics, University of Innsbruck, Innsbruck, Austria
| | - Lukas Schausberger
- Department of General and Surgical Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christine Schlömmer
- Department of General and Surgical Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Volker Schäfer
- Department of Anaesthesiology and Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Marlies Liebensteiner
- Department of General and Surgical Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Katharina Schäffler
- Department of General and Surgical Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Bettina Schenk
- Department of General and Surgical Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Fries
- Department of General and Surgical Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Petra Innerhofer
- Department of Anaesthesiology and Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Niederwanger
- Department of Pediatrics, Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
24
|
Iwako H, Tashiro H, Okimoto S, Yamaguchi M, Abe T, Kuroda S, Kobayashi T, Ohdan H. Antithrombin Insufficiency Promotes Susceptibility to Liver Tumorigenesis. J Surg Res 2018; 236:198-208. [PMID: 30694755 DOI: 10.1016/j.jss.2018.11.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/16/2018] [Accepted: 11/19/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND Antithrombin (AT) is not only a major regulator of hemostasis, but it shows anti-inflammatory properties as well. We aimed to investigate whether AT-insufficient mice increase susceptibility to liver tumorigenesis. METHODS We induced the development of liver tumor in AT-insufficient (AT+/-) mice and wild-type (AT+/+) mice by treating them with diethylnitrosamine (DEN) and CCl4. The development of liver tumors and liver inflammation were compared between these mouse groups. Following this, AT was administered to the AT-insufficient mice treated with DEN and CCl4. RESULTS Tumor size and the number of DEN and CCl4-induced liver tumors significantly increased in AT-insufficient mice compared with the wild-type mice. Serum transaminase levels, cell death, and the expression of cleaved caspase-3 in liver were increased in AT+/-. Furthermore, hepatic neutrophil infiltrations and serum interleukin 6 and tumor necrosis factor-α levels were significantly elevated in AT-insufficient mice. The levels of 8-OHdG, oxidative DNA damage marker, in liver were significantly increased in AT-insufficient mice. Administration of AT led to a significant decrease in DEN- and CCl4-induced liver injury and inflammation in AT-insufficient mice, compared with the wild-type group. CONCLUSIONS AT insufficiency led to increased susceptibility to liver tumorigenesis by increasing hepatic inflammation.
Collapse
Affiliation(s)
- Hiroshi Iwako
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Japan; Department of Clinical Research, Kure Medical Center, National Hospital Organization, Japan.
| | - Hirotaka Tashiro
- Department of Surgery, Kure Medical Center and Chugoku Cancer Center, National Hospital Organization, Japan
| | - Sho Okimoto
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Megumi Yamaguchi
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Japan; Department of Clinical Research, Kure Medical Center, National Hospital Organization, Japan
| | - Tomoyuki Abe
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Shintaro Kuroda
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| |
Collapse
|
25
|
Dinarvand P, Yang L, Villoutreix BO, Rezaie AR. Expression and functional characterization of two natural heparin-binding site variants of antithrombin. J Thromb Haemost 2018; 16:330-341. [PMID: 29215785 PMCID: PMC5809256 DOI: 10.1111/jth.13920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Indexed: 02/02/2023]
Abstract
Essentials Heparin-binding site (HBS) variants of antithrombin (AT) are associated with thrombosis risk. HSB variants have, in general, normal progressive inhibitory activity but reduced heparin affinity. Thrombosis in HSB carriers has been primarily attributed to the loss of heparin cofactor activity. Results here demonstrate that HSB variants of AT also lack anti-inflammatory signaling functions. SUMMARY Background Several heparin-binding site (HBS) variants of antithrombin (AT) have been identified that predispose carriers to a higher incidence of thrombosis. Thrombosis in carriers of HBS variants has been primarily attributed to a loss in their heparin-dependent anticoagulant function. Objective The objective of this study was to determine whether HSB mutations affect the anti-inflammatory functions of variants. Methods Two HBS variants of AT (AT-I7N and AT-L99F), which are known to be associated with a higher incidence of thrombosis, were expressed in mammalian cells and purified to homogeneity. These variants were characterized by kinetic assays followed by analysis of their activities in established cellular and/or in vivo inflammatory models. The possible effects of mutations on AT structure were also evaluated by molecular modeling. Results The results indicated that, whereas progressive inhibitory activities of variants were minimally affected, their heparin affinity and inhibitory activity in the presence of heparin were markedly decreased. Unlike wild-type AT, neither AT variant was capable of inhibiting activation of nuclear factor-κB or downregulation of expression of cell adhesion molecules in response to lipopolysaccharide (LPS). Similarly, neither variant elicited barrier protective activity in response to LPS. Structural analysis suggested that the L99F substitution locally destabilizes AT structure. Conclusions It is concluded that the L99F mutation of AT is associated with destabilization of the serpin structure, and that the loss of anti-inflammatory signaling function of the HBS variants may also contribute to enhanced thrombosis in carriers of HBS mutations.
Collapse
Affiliation(s)
- Peyman Dinarvand
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis
| | - Likui Yang
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
| | - Bruno O. Villoutreix
- Inserm U973, Université Paris Diderot, Sorbonne Paris Cité, Molécules Thérapeutiques In Silico, Paris, France
| | - Alireza R. Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| |
Collapse
|
26
|
Yin J, Wang F, Kong Y, Wu R, Zhang G, Wang N, Wang L, Lu Z, Liang M. Antithrombin III prevents progression of chronic kidney disease following experimental ischaemic-reperfusion injury. J Cell Mol Med 2017; 21:3506-3514. [PMID: 28767184 PMCID: PMC5706518 DOI: 10.1111/jcmm.13261] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 04/30/2017] [Indexed: 12/27/2022] Open
Abstract
Acute kidney disease (AKI) leads to increased risk of progression to chronic kidney disease (CKD). Antithrombin III (ATIII) is a potent anticoagulant with anti-inflammatory properties, and we previously reported that insufficiencies of ATIII exacerbated renal ischaemia-reperfusion injury (IRI) in rats. In this study, we examined the characteristic of AKI-CKD transition in rats with two distinct AKI models. Based on our observation, left IRI plus right nephrectomy (NX-IRI) was used to determine whether ATIII had therapeutic effects in preventing CKD progression after AKI. It was observed that NX-IRI resulted in significant functional and histological damage at 5 weeks after NX-IRI compared with sham rats, which was mitigated by ATIII administration. Besides, we noticed that ATIII administration significantly reduced NX-IRI-induced interstitial fibrosis. Consistently, renal expression of collagen-1, α-smooth muscle actin and fibronectin were substantial diminished in ATIII-administered rats compared with un-treated NX-IRI rats. Furthermore, the beneficial effects of ATIII were accompanied with decreased M1-like macrophage recruitment and down-regulation of M1-like macrophage-dependent pro-inflammatory cytokines such as tumour necrosis factor α, inducible nitric oxide synthase and interleukin-1β, indicating that ATIII prevented AKI-CKD transition via inhibiting inflammation. Overall, ATIII shows potential as a therapeutic strategy for the prevention of CKD progression after AKI.
Collapse
Affiliation(s)
- Jianyong Yin
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Feng Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yiwei Kong
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Rui Wu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guangyuan Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ling Wang
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeyuan Lu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
27
|
Zanoni M, Aventurato ÍK, Hunter J, Sucupira MCA, Diaz RS. Uniquely altered transcripts are associated with immune preservation in HIV infection. PLoS One 2017; 12:e0169868. [PMID: 28350860 PMCID: PMC5370105 DOI: 10.1371/journal.pone.0169868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 12/23/2016] [Indexed: 01/13/2023] Open
Abstract
The mechanisms underlying host HIV control hold much promise in the search for a functional HIV cure. We investigated the host genomic signatures in elite controllers or rapid progressors following recent infection and the correlates of immune reconstitution during combination antiretroviral therapy. We characterized the HIV-specific longitudinal host transcriptional response of peripheral blood mononuclear cells from elite controllers, rapid progressors, immune responders and non-responders using a RT-qPCR array in a cohort of recently HIV-infected Brazilian individuals. The elite controllers expressed unique transcripts early in infection that were closely associated with specialized cross-presentation between XCR1+ DCs and antigen-specific CD8+ T cells (XCL1). The natural suppression of HIV was also associated with the highly functional co-expression of cytokines and chemokines (CCL2, TNF and IL-10) concomitant with the maintenance of important anti-inflammatory and anticoagulant properties (Antithrombin III). Immune responders exhibited exclusively upregulated mRNAs possibly related to stem cell mobilization before combination antiretroviral therapy (neutrophil elastase). Our longitudinal approach to gene expression permitted us to discover previously unrecognized determinants that contribute to natural or antiretroviral-mediated HIV-1 immune control.
Collapse
Affiliation(s)
- Michelle Zanoni
- Retrovirology Laboratory, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Ítalo Karmann Aventurato
- Retrovirology Laboratory, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - James Hunter
- Retrovirology Laboratory, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | | - Ricardo Sobhie Diaz
- Retrovirology Laboratory, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
- * E-mail:
| |
Collapse
|
28
|
Lu Z, Cheng D, Yin J, Wu R, Zhang G, Zhao Q, Wang N, Wang F, Liang M. Antithrombin III Protects Against Contrast-Induced Nephropathy. EBioMedicine 2017; 17:101-107. [PMID: 28219627 PMCID: PMC5360582 DOI: 10.1016/j.ebiom.2017.02.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 11/26/2022] Open
Abstract
We previously reported that insufficiency of antithrombin III (ATIII), the major anti-coagulation molecule in vivo, exacerbated renal ischemia-reperfusion injury in animal models and possibly humans. In the present study, we investigated the relationship between ATIII level and contrast induced nephropathy (CIN) in patients and examined therapeutic effect of ATIII on CIN in Sprague-Dawley rats. Patients with low ATIII activity presented a higher incidence of acute kidney injury (AKI) following coronary angiography. ATIII (500 μg/kg) was intravenously injected before or after the induction of AKI in rats. Our data demonstrated ATIII significantly attenuated the elevation of serum creatinine, blood urea nitrogen, and renal histological injury. The beneficial effects of ATIII were accompanied by diminished renal inflammatory response, oxidative stress, cell apoptosis and improved renal blood flow in rats. In conclusion, ATIII appears to attenuate CIN through inhibiting inflammation, oxidative stress, apoptosis and improving renal blood flow. ATIII administration may represent a promising strategy for the prevention and treatment of contrast-induced AKI.
Patients with low ATIII activity presented a higher incidence of acute kidney injury following coronary angiography. ATIII supplementation attenuated renal injury in animal models of contrast induced nephropathy. ATIII exerted renoprotective effect by inhibiting inflammation, oxidative stress, apoptosis and improving renal blood flow. Antithrombin III (ATIII), a potent anti-coagulation molecule in vivo, has been reported that it can exert reno-protective effects in ischemia-reperfusion model. Nevertheless, whether exogenous ATIII administration can protect against contrast induced nephropathy (CIN) in animal models remains unclear. This study revealed that ATIII administration has therapeutic effects against CIN in Sprague-Dawley Rats. Furthermore, the reno-protection conferred by ATIII might be mediated by inhibition of inflammation, oxidative stress, apoptosis and improving renal blood flow. ATIII supplementation represents a promising prophylactic and treatment strategies for contrast induced AKI.
Collapse
Affiliation(s)
- Zeyuan Lu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Dongsheng Cheng
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Jianyong Yin
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Rui Wu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Guangyuan Zhang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China.
| | - Qing Zhao
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Feng Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
29
|
Smits NC, Kobayashi T, Srivastava PK, Skopelja S, Ivy JA, Elwood DJ, Stan RV, Tsongalis GJ, Sellke FW, Gross PL, Cole MD, DeVries JT, Kaplan AV, Robb JF, Williams SM, Shworak NW. HS3ST1 genotype regulates antithrombin's inflammomodulatory tone and associates with atherosclerosis. Matrix Biol 2017; 63:69-90. [PMID: 28126521 DOI: 10.1016/j.matbio.2017.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 12/21/2022]
Abstract
The HS3ST1 gene controls endothelial cell production of HSAT+ - a form of heparan sulfate containing a specific pentasaccharide motif that binds the anticoagulant protein antithrombin (AT). HSAT+ has long been thought to act as an endogenous anticoagulant; however, coagulation was normal in Hs3st1-/- mice that have greatly reduced HSAT+ (HajMohammadi et al., 2003). This finding indicates that HSAT+ is not essential for AT's anticoagulant activity. To determine if HSAT+ is involved in AT's poorly understood inflammomodulatory activities, Hs3st1-/- and Hs3st1+/+ mice were subjected to a model of acute septic shock. Compared with Hs3st1+/+ mice, Hs3st1-/- mice were more susceptible to LPS-induced death due to an increased sensitivity to TNF. For Hs3st1+/+ mice, AT treatment reduced LPS-lethality, reduced leukocyte firm adhesion to endothelial cells, and dilated isolated coronary arterioles. Conversely, for Hs3st1-/- mice, AT induced the opposite effects. Thus, in the context of acute inflammation, HSAT+ selectively mediates AT's anti-inflammatory activity; in the absence of HSAT+, AT's pro-inflammatory effects predominate. To explore if the anti-inflammatory action of HSAT+ also protects against a chronic vascular-inflammatory disease, atherosclerosis, we conducted a human candidate-gene association study on >2000 coronary catheterization patients. Bioinformatic analysis of the HS3ST1 gene identified an intronic SNP, rs16881446, in a putative transcriptional regulatory region. The rs16881446G/G genotype independently associated with the severity of coronary artery disease and atherosclerotic cardiovascular events. In primary endothelial cells, the rs16881446G allele associated with reduced HS3ST1 expression. Together with the mouse data, this leads us to conclude that the HS3ST1 gene is required for AT's anti-inflammatory activity that appears to protect against acute and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Nicole C Smits
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Takashi Kobayashi
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Pratyaksh K Srivastava
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Sladjana Skopelja
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Julianne A Ivy
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Dustin J Elwood
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Radu V Stan
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Gregory J Tsongalis
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Brown Medical School, Providence, RI, USA
| | - Peter L Gross
- Department of Medicine, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael D Cole
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - James T DeVries
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Aaron V Kaplan
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - John F Robb
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Scott M Williams
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Nicholas W Shworak
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA; Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| |
Collapse
|
30
|
Ishikawa M, Yamashita H, Oka N, Ueda T, Kohama K, Nakao A, Kotani J. Antithrombin III improved neutrophil extracellular traps in lung after the onset of endotoxemia. J Surg Res 2016; 208:140-150. [PMID: 27993201 DOI: 10.1016/j.jss.2016.09.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/05/2016] [Accepted: 09/21/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND Coagulation and inflammation are closely linked during acute inflammatory conditions, such as sepsis. Antithrombin (AT) is an anticoagulant that also has anti-inflammatory activities. The effects of therapeutically administering AT III after the onset of endotoxemia or sepsis were not clear. Here, we studied the effects of administering AT III after inducing lethal endotoxemia in mice. METHODS Mice were injected intraperitoneally with lipopolysaccharide (LPS) to induce endotoxemia. AT III was administered 3 h later. We assessed survival and the severity of endotoxemia and quantified plasma cytokine levels and biochemical markers of liver and kidney function. In the lungs, we examined neutrophil accumulation, neutrophil extracellular traps, alveolar wall thickness, and chemokine (C-X-C motif) ligand 1 (cxcl-1), cxcl-2, and high mobility group box 1 expression. RESULTS Administering AT III reduced the severity and mortality of LPS-induced endotoxemia as indicated by 24-h survival of 84% of the mice that received LPS + AT III and only 53% of mice given LPS alone (P < 0.05). AT III treatment attenuated several changes induced in the lungs by endotoxemia including cxcl-2 mRNA expression, high mobility group box 1 protein expression, neutrophil accumulation, alveolar septal thickening, and neutrophil extracellular trap formation. AT III did not decrease plasma cytokine levels or plasma urea nitrogen levels that were upregulated as a result of LPS-induced endotoxemia. CONCLUSIONS Administration of AT III after the onset of endotoxemia improved outcomes in a mouse model. The attenuation of lung inflammation may have a large impact on mortality and morbidity. Because lung inflammation increases the likelihood of mortality from sepsis, AT III could be a useful agent in septic patients.
Collapse
Affiliation(s)
- Michiko Ishikawa
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Japan.
| | - Hayato Yamashita
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Nobuki Oka
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Takahiro Ueda
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Keisuke Kohama
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Japan; Senri Critical Care Medical Center, Saiseikai Senri Hospital, Osaka, Japan
| | - Atsunori Nakao
- Department of Emergency and Critical Care Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Joji Kotani
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
31
|
Enhanced P-selectin expression on platelet-a marker of platelet activation, in young patients with angiographically proven coronary artery disease. Mol Cell Biochem 2016; 419:125-33. [DOI: 10.1007/s11010-016-2756-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/21/2016] [Indexed: 01/19/2023]
|
32
|
Antithrombin controls tumor migration, invasion and angiogenesis by inhibition of enteropeptidase. Sci Rep 2016; 6:27544. [PMID: 27270881 PMCID: PMC4897635 DOI: 10.1038/srep27544] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/17/2016] [Indexed: 12/18/2022] Open
Abstract
Antithrombin is a key inhibitor of the coagulation cascade, but it may also function as an anti-inflammatory, anti-angiogenic, anti-viral and anti-apoptotic protein. Here, we report a novel function of antithrombin as a modulator of tumor cell migration and invasion. Antithrombin inhibited enteropeptidase on the membrane surface of HT-29, A549 and U-87 MG cells. The inhibitory process required the activation of antithrombin by heparin, and the reactive center loop and the heparin binding domain were essential. Surprisingly, antithrombin non-covalently inhibited enteropeptidase, revealing a novel mechanism of inhibition for this serpin. Moreover, as a consequence of this inhibition, antithrombin was cleaved, resulting in a molecule with anti-angiogenic properties that reduced vessel-like formation of endothelial cells. The addition of antithrombin and heparin to U-87 MG and A549 cells reduced motility in wound healing assays, inhibited the invasion in transwell assays and the degradation of a gelatin matrix mediated by invadopodia. These processes were controlled by enteropeptidase, as demonstrated by RNA interference experiments. Carcinoma cell xenografts in nude mice showed in vivo co-localization of enteropeptidase and antithrombin. Finally, treatment with heparin reduced experimental metastasis induced by HT29 cells in vivo. In conclusion, the inhibition of enteropeptidase by antithrombin may have a double anti-tumor effect through inhibiting a protease involved in metastasis and generating an anti-angiogenic molecule.
Collapse
|
33
|
Swanepoel AC, Nielsen VG, Pretorius E. Viscoelasticity and Ultrastructure in Coagulation and Inflammation: Two Diverse Techniques, One Conclusion. Inflammation 2015; 38:1707-26. [PMID: 25772112 DOI: 10.1007/s10753-015-0148-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The process of blood clotting has been studied for centuries. A synopsis of current knowledge pertaining to haemostasis and the blood components, including platelets and fibrin networks which are closely involved in coagulation, are discussed. Special emphasis is placed on tissue factor (TF), calcium and thrombin since these components have been implicated in both the coagulation process and inflammation. Analysis of platelets and fibrin morphology indicate that calcium, tissue factor and thrombin at concentrations used during viscoelastic analysis (with thromboelastography or TEG) bring about alterations in platelet and fibrin network ultrastructure, which is similar to that seen in inflammation. Scanning electron microscopy indicated that, when investigating platelet structure in disease, addition of TF, calcium or thrombin will mask disease-induced alterations associated with platelet activation. Therefore, washed platelets without any additives is preferred for morphological analysis. Furthermore, morphological and viscoelastic analysis confirmed that thrombin activation is the preferred method of fibrin activation when investigating fibrin network ultrastructure.
Collapse
Affiliation(s)
- Albe C Swanepoel
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag x323, Arcadia, 0007, South Africa,
| | | | | |
Collapse
|
34
|
Kang SS, Baik JE, Yang JS, Cho K, Yun CH, Han SH. Protein profiles in mucosal and systemic compartments in response to Vibrio cholerae in a mouse pulmonary infection model. Microb Pathog 2015; 86:10-7. [PMID: 26150210 DOI: 10.1016/j.micpath.2015.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 06/17/2015] [Accepted: 07/02/2015] [Indexed: 11/19/2022]
Abstract
We have recently shown that a mouse lung infection model resulting in acute pneumonia could be used for evaluating the protective immunity induced by mucosal vaccines against Vibrio cholerae. In order to gain insight and better understanding of the pathogenicity of V. cholerae infection, we identified and compared proteins induced by V. cholerae in nasal washes, bronchoalveolar lavages (BAL), and sera. Intranasal administration of V. cholerae increased the concentration of total proteins in nasal washes and BAL fluids, but not in sera. LTQ-Orbitrap hybrid Fourier transform mass spectrometry showed that cytoskeletal proteins, protease inhibitors and anti-inflammatory mediators were present in nasal washes from uninfected mice. The distinctly expressed proteins in nasal washes in response to V. cholerae mainly consisted of protease inhibitors, anti-inflammatory proteins, and anti-microbial proteins. A number of protease inhibitors and anti-inflammatory proteins were selectively expressed in BAL fluids from V. cholerae-infected mice, while cytoskeletal proteins and heat shock proteins were mainly observed in BAL fluids from uninfected mice. A large number of serum complements, protease inhibitors, and acute phase proteins were expressed in V. cholerae-infected mice. Collectively, these results suggest that intranasal administration of V. cholerae leading to acute pneumonia elicited alterations of protein profiles associated with immune homeostasis and host protection in both the mucosal and systemic compartments.
Collapse
Affiliation(s)
- Seok-Seong Kang
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 110-749, Republic of Korea
| | - Jung Eun Baik
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 110-749, Republic of Korea
| | - Jae Seung Yang
- Clinical Immunology Section, Laboratory Sciences Division, International Vaccine Institute, Seoul 151-919, Republic of Korea
| | - Kun Cho
- Division of Mass Spectrometry Research, Korea Basic Science Institute, Ochang 863-883, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 151-921, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul 110-749, Republic of Korea.
| |
Collapse
|
35
|
Liu R, Jiang W, Walkey CD, Chan WCW, Cohen Y. Prediction of nanoparticles-cell association based on corona proteins and physicochemical properties. NANOSCALE 2015; 7:9664-75. [PMID: 25959034 DOI: 10.1039/c5nr01537e] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cellular association of nanoparticles (NPs) in biological fluids is affected by proteins adsorbed onto the NP surface, forming a "protein corona", thereby impacting cellular bioactivity. Here we investigate, based on an extensive gold NPs protein corona dataset, the relationships between NP-cell association and protein corona fingerprints (PCFs) as well as NP physicochemical properties. Accordingly, quantitative structure-activity relationships (QSARs) were developed based on both linear and non-linear support vector regression (SVR) models making use of a sequential forward floating selection of descriptors. The SVR model with only 6 serum proteins and zeta potential had higher accuracy (R(2) = 0.895) relative to the linear model (R(2) = 0.850) with 11 PCFs. Considering the initial pool of 148 descriptors, the APOB, A1AT, ANT3, and PLMN serum proteins along with NP zeta potential were identified as most significant to correlating NP-cell association. The present study suggests that QSARs exploration of NP-cell association data, considering the role of both NP protein corona and physicochemical properties, can support the planning and interpretation of toxicity studies and guide the design of NPs for biomedical applications.
Collapse
Affiliation(s)
- Rong Liu
- Center for Environmental Implications of Nanotechnology, University of California, Los Angeles, CA90095, USA.
| | | | | | | | | |
Collapse
|
36
|
Giannotta M, Tapete G, Emmi G, Silvestri E, Milla M. Thrombosis in inflammatory bowel diseases: what's the link? Thromb J 2015; 13:14. [PMID: 25866483 PMCID: PMC4393581 DOI: 10.1186/s12959-015-0044-2] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/26/2015] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease affects more than 2 million people in Europe, with almost 20% of patients being diagnosed in pediatric age. Patients with inflammatory bowel disease are at increased risk of thromboembolic complications which may affect patients’ morbidity and mortality. The risk of the most common thromboembolic events, such as deep venous thrombosis and pulmonary embolism, are estimated to be three-fold increased compared to controls, but many other districts can be affected. Moreover, patients with ulcerative colitis and Crohn’s disease experience thromboembolic events at a younger age compared to general population. Many factors have been investigated as determinants of the pro-thrombotic tendency such as acquired risk factors or genetic and immune abnormalities, but a unique cause has not been found. Many efforts have been focused on the study of abnormalities in the coagulation cascade, its natural inhibitors and the fibrinolytic system components and both quantitative and qualitative alterations have been demonstrated. Recently the role of platelets and microvascular endothelium has been reviewed, as the possible link between the inflammatory and hemostatic process.
Collapse
Affiliation(s)
- Martina Giannotta
- Gastroenterology Department, AOU Careggi Regional Referral Center for Inflammatory Bowel Disease, Florence, Italy
| | - Gherardo Tapete
- Gastroenterology Department, AOU Careggi Regional Referral Center for Inflammatory Bowel Disease, Florence, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Florence and Patologia Medica Unit, AOU Careggi, Florence, Italy
| | - Elena Silvestri
- Department of Experimental and Clinical Medicine, University of Florence and Patologia Medica Unit, AOU Careggi, Florence, Italy
| | - Monica Milla
- Gastroenterology Department, AOU Careggi Regional Referral Center for Inflammatory Bowel Disease, Florence, Italy
| |
Collapse
|
37
|
Senchenkova EY, Russell J, Esmon CT, Granger DN. Roles of Coagulation and fibrinolysis in angiotensin II-enhanced microvascular thrombosis. Microcirculation 2015; 21:401-7. [PMID: 24495184 DOI: 10.1111/micc.12120] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 01/29/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE AngII-induced HTN is associated with accelerated thrombus development in arterioles. This study assessed the contributions of different components of the coagulation cascade and fibrinolysis to AngII-mediated microvascular thrombosis. METHODS Light/dye-induced thrombus formation (the time of onset and flow cessation) was quantified in cremaster muscle arterioles of AngII infused (two weeks) WT/AngII mice, EPCR-TgN, and mice deficient in PAI-1. WT/AngII mice were also treated with either tissue factor antibody, antithrombin III, heparin, hirudin, or murine APC. RESULTS TF immunoblockade or hirudin treatment did not prevent the AngII-induced acceleration of thrombosis. While antithrombin III treatment prevented the acceleration in both thrombus onset and flow cessation, heparin only improved the time for blood flow cessation. Neither WT mice treated with murine APC nor EPCR-TgN were protected against AngII-induced thrombus development. A similar lack of protection was noted in PAI-1deficient mice. CONCLUSION These findings implicate a role for thrombin generation pathway in the accelerated thrombosis induced by AngII and suggest that an impaired protein C pathway and increased PAI-1 do not make a significant contribution to this model of microvascular thrombosis.
Collapse
Affiliation(s)
- Elena Y Senchenkova
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, Louisiana, USA; Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint-Petersburg, Russia
| | | | | | | |
Collapse
|
38
|
Coagulation abnormalities in sepsis. ACTA ACUST UNITED AC 2015; 53:16-22. [DOI: 10.1016/j.aat.2014.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 11/16/2014] [Accepted: 11/24/2014] [Indexed: 11/30/2022]
|
39
|
Papareddy P, Kalle M, Bhongir RKV, Mörgelin M, Malmsten M, Schmidtchen A. Antimicrobial effects of helix D-derived peptides of human antithrombin III. J Biol Chem 2014; 289:29790-800. [PMID: 25202017 DOI: 10.1074/jbc.m114.570465] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Antithrombin III (ATIII) is a key antiproteinase involved in blood coagulation. Previous investigations have shown that ATIII is degraded by Staphylococcus aureus V8 protease, leading to release of heparin binding fragments derived from its D helix. As heparin binding and antimicrobial activity of peptides frequently overlap, we here set out to explore possible antibacterial effects of intact and degraded ATIII. In contrast to intact ATIII, the results showed that extensive degradation of the molecule yielded fragments with antimicrobial activity. Correspondingly, the heparin-binding, helix D-derived, peptide FFFAKLNCRLYRKANKSSKLV (FFF21) of human ATIII, was found to be antimicrobial against particularly the Gram-negative bacteria Escherichia coli and Pseudomonas aeruginosa. Fluorescence microscopy and electron microscopy studies demonstrated that FFF21 binds to and permeabilizes bacterial membranes. Analogously, FFF21 was found to induce membrane leakage of model anionic liposomes. In vivo, FFF21 significantly reduced P. aeruginosa infection in mice. Additionally, FFF21 displayed anti-endotoxic effects in vitro. Taken together, our results suggest novel roles for ATIII-derived peptide fragments in host defense.
Collapse
Affiliation(s)
- Praveen Papareddy
- From the Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Biomedical Center, Tornavägen 10, SE-221 84 Lund, Sweden, the Division of Infection Medicine, Department of Clinical Sciences, Lund University, Biomedical Center, Tornavägen 10, SE-221 84 Lund, Sweden,
| | - Martina Kalle
- From the Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Biomedical Center, Tornavägen 10, SE-221 84 Lund, Sweden
| | - Ravi K V Bhongir
- the Division of Infection Medicine, Department of Clinical Sciences, Lund University, Biomedical Center, Tornavägen 10, SE-221 84 Lund, Sweden
| | - Matthias Mörgelin
- the Division of Infection Medicine, Department of Clinical Sciences, Lund University, Biomedical Center, Tornavägen 10, SE-221 84 Lund, Sweden
| | - Martin Malmsten
- the Department of Pharmacy, Uppsala University, SE-751 23, Uppsala, Sweden, and
| | - Artur Schmidtchen
- From the Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Biomedical Center, Tornavägen 10, SE-221 84 Lund, Sweden, the LKCMedicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232
| |
Collapse
|
40
|
Antithrombin attenuates vascular leakage via inhibiting neutrophil activation in acute lung injury. Crit Care Med 2014; 41:e439-46. [PMID: 24107637 DOI: 10.1097/ccm.0b013e318298ad3a] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To test the hypothesis that restoration of antithrombin plasma concentrations attenuates vascular leakage by inhibiting neutrophil activation through syndecan-4 receptor inhibition in an established ovine model of acute lung injury. DESIGN Randomized controlled laboratory experiment. SETTING University animal research facility. SUBJECTS Eighteen chronically instrumented sheep. INTERVENTIONS Following combined burn and smoke inhalation injury (40% of total body surface area, third-degree flame burn; 4 × 12 breaths of cold cotton smoke), chronically instrumented sheep were randomly assigned to receive an IV infusion of 6 IU/kg/hr recombinant human antithrombin III or normal saline (n = 6 each) during the 48-hour study period. In addition, six sham animals (not injured, continuous infusion of vehicle) were used to obtain reference values for histological and immunohistochemical analyses. MEASUREMENTS AND MAIN RESULTS Compared to control animals, recombinant human antithrombin III reduced the number of neutrophils per hour in the pulmonary lymph (p < 0.01 at 24 and 48 hr), alveolar neutrophil infiltration (p = 0.04), and pulmonary myeloperoxidase activity (p = 0.026). Flow cytometric analysis revealed a significant reduction of syndecan-4-positive neutrophils (p = 0.002 vs control at 24 hr). Treatment with recombinant human antithrombin III resulted in a reduction of pulmonary nitrosative stress (p = 0.002), airway obstruction (bronchi: p = 0.001, bronchioli: p = 0.013), parenchymal edema (p = 0.044), and lung bloodless wet-to-dry-weight ratio (p = 0.015). Clinically, recombinant human antithrombin III attenuated the increased pulmonary transvascular fluid flux (12-48 hr: p ≤ 0.001 vs control each) and the deteriorated pulmonary gas exchange (12-48 hr: p < 0.05 vs control each) without increasing the risk of bleeding. CONCLUSIONS The present study provides evidence for the interaction between antithrombin and neutrophils in vivo, its pathophysiological role in vascular leakage, and the therapeutic potential of recombinant human antithrombin III in a large animal model of acute lung injury.
Collapse
|
41
|
Long N, Ng S, Donnelly G, Owens M, McNicholas M, McCarthy K, McCaul C. Anatomical characterisation of the cricothyroid membrane in females of childbearing age using computed tomography. Int J Obstet Anesth 2013; 23:10-7. [PMID: 24291169 DOI: 10.1016/j.ijoa.2013.07.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 07/05/2013] [Accepted: 07/06/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND In the event of failure to secure the airway by conventional means, it may be necessary to perform invasive airway access via the cricothyroid membrane. No studies have addressed anatomy of this structure in the obstetric population. We aimed to review the anatomical variation of this structure in a population of childbearing age. METHODS We searched the radiology database for computed tomography studies of the neck performed in a 13-month period in consecutive patients aged 15-55 years. Studies on 18 females and 22 males were reviewed. Male patients were included for comparison. Data were reconstructed using a high spatial frequency algorithm to optimise spatial resolution. Five parameters were measured: distance from the skin to the membrane, maximum midline height of the membrane in the vertical plane, maximum transverse diameter of the membrane, neck diameter and cartilaginous calcification. RESULTS The distance (mean range) from skin to the membrane was similar in females and males (16.2 [3-33] vs. 13.9 [3-37] mm, P = 0.42). The vertical height (9.9 [7-17] vs. 11.4 [8-15] mm, P = 0.04) and maximum width of the membrane (14.5 [10-17] mm vs. 12.5 [10-15] mm, P < 0.01) were greater in males. Cartilaginous calcification was low and did not differ between genders. CONCLUSIONS The cricothyroid membrane is not necessarily a superficial structure and consequently may be difficult to palpate. The smallest dimensions of the membrane indicate that smaller than recommended cricothyroidotomy devices may be required in some patients as the external diameter of commercial trocar devices and tracheal tubes may exceed 7 mm.
Collapse
Affiliation(s)
- N Long
- Department of Radiology, The Mater Misericordiae University Hospital, Dublin, Ireland
| | - S Ng
- Department of Anaesthesia, The Rotunda Hospital, Dublin, Ireland
| | - G Donnelly
- Department of Anaesthesia, The Rotunda Hospital, Dublin, Ireland; Department of Anaesthesia, Mater Misericordiae, University Hospital, Dublin, Ireland
| | - M Owens
- Department of Anaesthesia, Mater Misericordiae, University Hospital, Dublin, Ireland
| | - M McNicholas
- Department of Radiology, The Mater Misericordiae University Hospital, Dublin, Ireland
| | - K McCarthy
- Department of Anaesthesia, The Rotunda Hospital, Dublin, Ireland
| | - C McCaul
- Department of Anaesthesia, The Rotunda Hospital, Dublin, Ireland; Department of Anaesthesia, Mater Misericordiae, University Hospital, Dublin, Ireland; School of Medicine and Medical Sciences, University College Dublin, Ireland.
| |
Collapse
|
42
|
The role of the vessel wall. Methods Mol Biol 2013; 992:31-46. [PMID: 23546703 DOI: 10.1007/978-1-62703-339-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The role of the vessel wall is complex and its effects are wide-ranging. The vessel wall, specifically the endothelial monolayer that lines the inner lumen, possesses the ability to influence various physiological states both locally and systemically by controlling vascular tone, basement membrane component synthesis, angiogenesis, haemostatic properties, and immunogenicity. This is an overview of the function and structure of the vessel wall and how disruption and dysfunction in any of these regulatory roles can lead to disease states.
Collapse
|
43
|
Menschikowski M, Hagelgans A, Schuler U, Froeschke S, Rosner A, Siegert G. Plasma Levels of Phospholipase A2-IIA in Patients with Different Types of Malignancies: Prognosis and Association with Inflammatory and Coagulation Biomarkers. Pathol Oncol Res 2013; 19:839-46. [DOI: 10.1007/s12253-013-9652-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 05/05/2013] [Indexed: 12/13/2022]
|
44
|
Antithrombin Reduces Inflammation and Microcirculatory Perfusion Failure in Closed Soft-Tissue Injury and Endotoxemia. Crit Care Med 2013; 41:867-73. [DOI: 10.1097/ccm.0b013e3182742d2c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
45
|
O'Brien M. The reciprocal relationship between inflammation and coagulation. Top Companion Anim Med 2012; 27:46-52. [PMID: 23031455 DOI: 10.1053/j.tcam.2012.06.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 06/18/2012] [Indexed: 12/21/2022]
Abstract
Inflammation and coagulation constitute two host defense systems with complementary roles in eliminating invading pathogens, limiting tissue damage, and restoring homeostasis. Extensive cross talk exists between these 2 systems, whereby inflammation leads to activation of coagulation, and coagulation considerably affects inflammatory activity. Infection leads to the production of proinflammatory cytokines that, in turn, stimulate the production of tissue factor. Activation of the coagulation system and ensuing thrombin generation are dependent on the expression of tissue factor. Conversely, activated coagulation proteases may affect specific receptors on inflammatory cells and endothelial cells and thereby modulate the inflammatory response. Activation of coagulation with the simultaneous down-regulation of endothelial-bound anticoagulant mechanisms and endogenous fibrinolysis characterizes the pathophysiology of sepsis. The mechanisms by which these highly complex and codependent defense strategies are linked together both in health and disease is the focus of this review.
Collapse
Affiliation(s)
- Mauria O'Brien
- University of Illinois Urbana-Champaign, College of Veterinary Medicine, Urbana, IL 61802, USA.
| |
Collapse
|
46
|
Prevention of High-Mobility Group Box 1-Mediated Early Loss of Transplanted Mouse Islets in the Liver by Antithrombin III. Transplantation 2012; 93:983-8. [DOI: 10.1097/tp.0b013e31824d3508] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
47
|
Miyazaki M, Kato M, Tanaka M, Tanaka K, Takao S, Kohjima M, Ito T, Enjoji M, Nakamuta M, Kotoh K, Takayanagi R. Antithrombin III injection via the portal vein suppresses liver damage. World J Gastroenterol 2012; 18:1884-91. [PMID: 22563168 PMCID: PMC3337563 DOI: 10.3748/wjg.v18.i16.1884] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 12/20/2011] [Accepted: 12/31/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of antithrombin III (AT III) injection via the portal vein in acute liver failure.
METHODS: Thirty rats were intraperitoneally challenged with lipopolysaccharide (LPS) and D-galactosamine (GalN) and divided into three groups: a control group; a group injected with AT III via the tail vein; and a group injected with AT III via the portal vein. AT III (50 U/kg body weight) was administrated 1 h after challenge with LPS and GalN. Serum levels of inflammatory cytokines and fibrin degradation products, hepatic fibrin deposition, and hepatic mRNA expression of hypoxia-related genes were analyzed.
RESULTS: Serum levels of alanine aminotransferase, tumor necrosis factor-α and interleukin-6 decreased significantly following portal vein AT III injection compared with tail vein injection, and control rats. Portal vein AT III injection reduced liver cell destruction and decreased hepatic fibrin deposition. This treatment also significantly reduced hepatic mRNA expression of lactate dehydrogenase and heme oxygenase-1.
CONCLUSION: A clinically acceptable dose of AT III injection into the portal vein suppressed liver damage, probably through its enhanced anticoagulant and anti-inflammatory activities.
Collapse
|
48
|
Aslami H, Haitsma JJ, Hofstra JJ, Florquin S, Dos Santos C, Streutker C, Zhang H, Levi M, Slutsky AS, Schultz MJ. Plasma-derived human antithrombin attenuates ventilator-induced coagulopathy but not inflammation in a Streptococcus pneumoniae pneumonia model in rats. J Thromb Haemost 2012; 10:399-410. [PMID: 22236057 DOI: 10.1111/j.1538-7836.2012.04622.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Mechanical ventilation exaggerates pneumonia-associated pulmonary coagulopathy and inflammation. We hypothesized that the administration of plasma-derived human antithrombin (AT), one of the natural inhibitors of coagulation, prevents ventilator-induced pulmonary coagulopathy, inflammation and bacterial outgrowth in a Streptococcus pneumoniae pneumonia model in rats. METHODS Forty-eight hours after induction of S. pneumoniae pneumonia rats were subjected to mechanical ventilation (tidal volume 12 mL kg(-1), positive end-expiratory pressure 0 cmH(2)O and inspired oxygen fraction 40%). Rats were randomized to systemic treatment with AT (250 IU administered intravenously (i.v.) before the start of mechanical ventilation) or placebo (saline). Non-ventilated, non-infected rats and non-ventilated rats with pneumonia served as controls. The primary endpoints were pulmonary coagulation and inflammation in bronchoalveolar lavage fluid (BALF). RESULTS Pneumonia was characterized by local activation of coagulation and inhibition of fibrinolysis, resulting in increased levels of fibrin degradation products and fibrin deposition in the lung. Mechanical ventilation exaggerated pulmonary coagulopathy and inflammation. Systemic administration of AT led to supra-normal BALF levels of AT and decreased ventilator-associated activation of coagulation. AT neither affected pulmonary inflammation nor bacterial outgrowth from the lungs or blood. CONCLUSIONS Plasma-derived human AT attenuates ventilator-induced coagulopathy, but not inflammation and bacterial outgrowth in a S. pneumoniae pneumonia model in rats.
Collapse
Affiliation(s)
- H Aslami
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hook KM, Abrams CS. The loss of homeostasis in hemostasis: new approaches in treating and understanding acute disseminated intravascular coagulation in critically ill patients. Clin Transl Sci 2012; 5:85-92. [PMID: 22376264 PMCID: PMC5439915 DOI: 10.1111/j.1752-8062.2011.00351.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Disseminated intravascular coagulation (DIC) profoundly increases the morbidity and mortality of patients who have sepsis. Both laboratory and clinical research advanced the understanding of the biology and pathophysiology of DIC. This, in turn, gave rise to improved therapies and patient outcomes. Beginning with a stimulus causing disruption of vascular integrity, cytokines and chemokines cause activation of systemic coagulation and inflammation. Seemingly paradoxically, the interplay between coagulation and inflammation also inhibits endogenous anticoagulants, fibrinolytics, and antiinflammatory pathways. The earliest documented and best-studied microbial cause of DIC is the lipopolysaccharide endotoxin of Gram-negative bacteria. Extensive microvascular thrombi emerge in the systemic vasculature due to dysregulation of coagulation. The result of this unrestrained, widespread small vessel thromboses multiorgan system failure. Consumption of platelets and coagulation factors during this process can lead to an elevated risk of hemorrhage. The management of these patients with simultaneous hemorrhage and thrombosis is complex and challenging. Definitive treatment of DIC, and attenuation of end-organ damage, requires control of the inciting cause. Currently, activated protein C is the only approved therapy in the United States for sepsis complicated by DIC. Further research is needed in this area to improve clinical outcomes for patients with sepsis.
Collapse
Affiliation(s)
- Karen M Hook
- Division of Hematology/Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| | | |
Collapse
|
50
|
Burgener A, Rahman S, Ahmad R, Lajoie J, Ramdahin S, Mesa C, Brunet S, Wachihi C, Kimani J, Fowke K, Carr S, Plummer F, Ball TB. Comprehensive proteomic study identifies serpin and cystatin antiproteases as novel correlates of HIV-1 resistance in the cervicovaginal mucosa of female sex workers. J Proteome Res 2011; 10:5139-49. [PMID: 21973077 DOI: 10.1021/pr200596r] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Not all individuals exposed to HIV-1 become infected, and evidence from HIV-1 highly exposed seronegative women (HIV-1-resistant) suggests that mucosal factors in the female genital tract, the first site of contact for the virus, are playing a role. To better understand factors mediating protection from HIV-1, we performed a large clinical study using the tools of systems biology to fully characterize the cervicovaginal mucosa proteome in HIV-1-resistant women. Cervicovaginal lavage fluid was collected from 293 HIV-1-resistant, uninfected, and infected sex workers and analyzed by 2D-LC LTQ-FT-MS. Of the more than 360 unique proteins identified, 41 were differentially abundant (>3-fold cutoff) in HIV-1-resistant women. The majority of over-abundant proteins were antiproteases (>40%), some with described anti-inflammatory and anti-HIV-1 activity. Quantification of specific anti-HIV-1 antiproteases Serpin A1, Serpin A3, and Cystatin B and an epithelial antiprotease A2ML1 found them to be significantly over-abundant in HIV-1-resistant women (p = 0.004; p = 0.046; p = 0.0003; and p = 0.04, respectively). Expression levels were not correlated to sexual practices or other epidemiological factors. Mucosal antiprotease levels correlated with pro-inflammatory cytokine concentration (p = <0.0001), but independently of pro-inflammatory cytokine levels in HIV-1-resistant women including TNF-alpha, IL-1 alpha, IL-1 beta, IL-6, and IL-8. This comprehensive systems biology approach identifies mucosal serpins and cystatins as novel correlates of HIV-1-resistance. This represents the first study characterizing these factors in the female genital tract.
Collapse
Affiliation(s)
- A Burgener
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|