1
|
Fan L, Tang K, Li J, Tan Y, Liu X, Bai Z, Tao A, Tan N. Mailuoning oral liquid ameliorates vasculitis in thromboangiitis obliterans rats via inactivating cGAS-STING-IRF3 and TLR4-MAPKs/NF-κB signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118707. [PMID: 39181282 DOI: 10.1016/j.jep.2024.118707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/07/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mailuoning oral liquid (MLN O), one traditional Chinese patent medicine, has a good therapeutic effect on thromboangiitis obliterans (TAO) in clinical practice. However, the underlying mechanism remains unclear. AIM OF THE STUDY This study aimed to explore the effects and potential mechanisms of MLN O against TAO based on network pharmacology and experimental verification. MATERIALS AND METHODS Network pharmacology was used to identify the intersectional targets and signaling pathways of MLN O and TAO. In vivo, the TAO model was established by injecting sodium laurate and dihydrotestosterone (DHT) into the femoral arteries of Wistar rats. Rats were given the indicated drugs by intragastric administration (i.g.), intravenous injection (i.v.), or subcutaneous injection (s.c.) per day for 21 days since a week before surgery. In vitro, HUVECs, RAW264.7, and THP-1 cells were stimulated by LPS and DHT to simulate the pathological changes of TAO. The anti-inflammatory, anticoagulant, and immunomodulatory effects of MLN O were evaluated by histological observation, blood biochemical indexes detection, H&E staining, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, western blotting and immunofluorescence assays. Furthermore, the vascular ring test was applied to explore the vasodilatory activity of MLN O. RESULTS MLN O significantly improved the pathological signs in TAO rats through its excellent anti-inflammatory, anticoagulant, immunomodulatory, and vasodilatory effects. Specifically, MLN O alleviated the gangrene and reduced the thrombosis in TAO rats, meanwhile, suppressed the expressions of inflammatory factors and clotting factors, which is related to the inactivations of cGAS-STING-IRF3 and TLR4-MAPKs/NF-κB signaling pathways. However, the superphysiological dose of DHT deteriorated the pathological development of TAO in vitro and in vivo. Moreover, the results of network pharmacology are consistent with the experimental verification. CONCLUSION Collectively, this study indicates for the first time that MLN O could alleviate TAO by inhibiting cGAS-STING-IRF3 and TLR4-MAPKs/NF-κB signaling pathways, which sheds light on a novel clinical therapeutic strategy for TAO.
Collapse
Affiliation(s)
- Lingling Fan
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Kai Tang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jian Li
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Jinling Pharmaceutical Co., Ltd., Nanjing, 210009, China
| | - Yajie Tan
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoqiong Liu
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Ziyu Bai
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Anhua Tao
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Ninghua Tan
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
2
|
Jiang J, Zhang X, Wang H, Spanos M, Jiang F, Ni L, Li J, Li G, Lin Y, Xiao J. Closer to The Heart: Harnessing the Power of Targeted Extracellular Vesicle Therapies. Adv Biol (Weinh) 2024; 8:e2300141. [PMID: 37953665 DOI: 10.1002/adbi.202300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/08/2023] [Indexed: 11/14/2023]
Abstract
Extracellular vesicles (EVs) have emerged as novel diagnostic and therapeutic approaches for cardiovascular diseases. EVs derived from various origins exhibit distinct effects on the cardiovascular system. However, the application of native EVs is constrained due to their poor stabilities and limited targeting capabilities. Currently, targeted modification of EVs primarily involves genetic engineering, chemical modification (covalent, non-covalent), cell membrane modification, and biomaterial encapsulation. These techniques enhance the stability, biological activity, target-binding capacity, and controlled release of EVs at specific cells and tissues. The diverse origins of cardioprotective EVs are covered, and the applications of cardiac-targeting EV delivery systems in protecting against cardiovascular diseases are discussed. This review summarizes the current stage of research on the potential of EV-based targeted therapies for addressing cardiovascular disorders.
Collapse
Affiliation(s)
- Jizong Jiang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xinxin Zhang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Hongyun Wang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Fei Jiang
- Department of Nursing, Union Hospital, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Lingyan Ni
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jin Li
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yanjuan Lin
- Department of Nursing, Union Hospital, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
3
|
Fathieh S, Grieve SM, Negishi K, Figtree GA. Potential Biological Mediators of Myocardial and Vascular Complications of Air Pollution-A State-of-the-Art Review. Heart Lung Circ 2023; 32:26-42. [PMID: 36585310 DOI: 10.1016/j.hlc.2022.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 12/29/2022]
Abstract
Ambient air pollution is recognised globally as a significant contributor to the burden of cardiovascular diseases. The evidence from both human and animal studies supporting the cardiovascular impact of exposure to air pollution has grown substantially, implicating numerous pathophysiological pathways and related signalling mediators. In this review, we summarise the list of activated mediators for each pathway that lead to myocardial and vascular injury in response to air pollutants. We performed a systematic search of multiple databases, including articles between 1990 and Jan 2022, summarising the evidence for activated pathways in response to each significant air pollutant. Particulate matter <2.5 μm (PM2.5) was the most studied pollutant, followed by particulate matter between 2.5 μm-10 μm (PM10), nitrogen dioxide (NO2) and ozone (O3). Key pathogenic pathways that emerged included activation of systemic and local inflammation, oxidative stress, endothelial dysfunction, and autonomic dysfunction. We looked at how potential mediators of each of these pathways were linked to both cardiovascular disease and air pollution and included the overlapping mediators. This review illustrates the complex relationship between air pollution and cardiovascular diseases, and discusses challenges in moving beyond associations, towards understanding causal contributions of specific pathways and markers that may inform us regarding an individual's exposure, response, and likely risk.
Collapse
Affiliation(s)
- Sina Fathieh
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Stuart M Grieve
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Radiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Kazuaki Negishi
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tas, Australia; Department of Cardiology, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan; Sydney Medical School Nepean, Faculty of Medicine and Health, Charles Perkins Centre Nepean, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Nepean Hospital, Sydney, NSW, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia.
| |
Collapse
|
4
|
LncRNA SOX2OT facilitates LPS-induced inflammatory injury by regulating intercellular adhesion molecule 1 (ICAM1) via sponging miR-215-5p. Clin Immunol 2022; 238:109006. [DOI: 10.1016/j.clim.2022.109006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/07/2022] [Accepted: 04/06/2022] [Indexed: 11/20/2022]
|
5
|
Phillippi DT, Daniel S, Pusadkar V, Youngblood VL, Nguyen KN, Azad RK, McFarlin BK, Lund AK. Inhaled diesel exhaust particles result in microbiome-related systemic inflammation and altered cardiovascular disease biomarkers in C57Bl/6 male mice. Part Fibre Toxicol 2022; 19:10. [PMID: 35135577 PMCID: PMC8827295 DOI: 10.1186/s12989-022-00452-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The gut microbiota plays a vital role in host homeostasis and is associated with inflammation and cardiovascular disease (CVD) risk. Exposure to particulate matter (PM) is a known mediator of inflammation and CVD and is reported to promote dysbiosis and decreased intestinal integrity. However, the role of inhaled traffic-generated PM on the gut microbiome and its corresponding systemic effects are not well-characterized. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) alters the gut microbiome and promotes microbial-related inflammation and CVD biomarkers. 4-6-week-old male C57Bl/6 mice on either a low-fat (LF, 10% fat) or high-fat (HF, 45% fat) diet were exposed via oropharyngeal aspiration to 35 μg DEP suspended in 35 μl saline or saline only (CON) 2x/week for 30 days. To determine whether probiotics could prevent diet or DEP exposure mediated alterations in the gut microbiome or systemic outcomes, a subset of animals on the HF diet were treated orally with 0.3 g/day (~ 7.5 × 108 CFU/day) of Winclove Ecologic® Barrier probiotics throughout the study. RESULTS Our results show that inhaled DEP exposure alters gut microbial profiles, including reducing Actinobacteria and expanding Verrucomicrobia and Proteobacteria. We observed increased circulating LPS, altered circulating cytokines (IL-1α, IL-3, IL-13, IL-15, G-CSF, LIF, MIP-2, and TNF-α), and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in DEP-exposed and/or HF diet mice. Furthermore, probiotics attenuated the observed reduction of Actinobacteria and expansion of Proteobacteria in DEP-exposed and HF-diet mice. Probiotics mitigated circulating cytokines (IL-3, IL-13, G-CSF, RANTES, and TNF- α) and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in respect to DEP-exposure and/or HF diet. CONCLUSION Key findings of this study are that inhaled DEP exposure alters small intestinal microbial profiles that play a role in systemic inflammation and early CVD biomarkers. Probiotic treatment in this study was fundamental in understanding the role of inhaled DEP on the microbiome and related systemic inflammatory and CVD biomarkers.
Collapse
Affiliation(s)
- Danielle T. Phillippi
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Sarah Daniel
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Vaidehi Pusadkar
- BioDiscovery Institute, Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
| | - Victoria L. Youngblood
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Kayla N. Nguyen
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Rajeev K. Azad
- BioDiscovery Institute, Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
- Department of Mathematics, University of North Texas, Denton, TX 76203 USA
| | - Brian K. McFarlin
- Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
- UNT Applied Physiology Laboratory, University of North Texas, Denton, TX 76203 USA
| | - Amie K. Lund
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| |
Collapse
|
6
|
Muñoz‐Ortiz T, Hu J, Ortgies DH, Shrikhande S, Zamora‐Perez P, Granado M, González‐Hedström D, Fuente‐Fernández M, García‐Villalón ÁL, Andrés‐Delgado L, Martín Rodríguez E, Aguilar R, Alfonso F, García Solé J, Rivera Gil P, Jaque D, Rivero F. Molecular Imaging of Infarcted Heart by Biofunctionalized Gold Nanoshells. Adv Healthc Mater 2021; 10:e2002186. [PMID: 33594792 DOI: 10.1002/adhm.202002186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Indexed: 01/03/2023]
Abstract
The unique combination of physical and optical properties of silica (core)/gold (shell) nanoparticles (gold nanoshells) makes them especially suitable for biomedicine. Gold nanoshells are used from high-resolution in vivo imaging to in vivo photothermal tumor treatment. Furthermore, their large scattering cross-section in the second biological window (1000-1700 nm) makes them also especially adequate for molecular optical coherence tomography (OCT). In this work, it is demonstrated that, after suitable functionalization, gold nanoshells in combination with clinical OCT systems are capable of imaging damage in the myocardium following an infarct. Since both inflammation and apoptosis are two of the main mechanisms underlying myocardial damage after ischemia, such damage imaging is achieved by endowing gold nanoshells with selective affinity for the inflammatory marker intercellular adhesion molecule 1 (ICAM-1), and the apoptotic marker phosphatidylserine. The results here presented constitute a first step toward a fast, safe, and accurate diagnosis of damaged tissue within infarcted hearts at the molecular level by means of the highly sensitive OCT interferometric technique.
Collapse
Affiliation(s)
- Tamara Muñoz‐Ortiz
- Nanomaterials for Bioimaging Group Departamento de Física de Materiales Universidad Autónoma de Madrid C/ Francisco Tomás y Valiente 7 Madrid 28049 Spain
| | - Jie Hu
- Xiamen Institute of Rare‐earth Materials, Haixi Institutes Chinese Academy of Sciences 258 Duishanxiheng Road, Jimei District Xiamen Fujian 361024 China
| | - Dirk H. Ortgies
- Nanomaterials for Bioimaging Group Departamento de Física de Materiales Universidad Autónoma de Madrid C/ Francisco Tomás y Valiente 7 Madrid 28049 Spain
- Instituto Ramón y Cajal de Investigación Sanitaria Hospital Ramón y Cajal Ctra. Colmenar km. 9,100 Madrid 28034 Spain
| | - Shreya Shrikhande
- Integrative Biomedical Materials and Nanomedicine Lab Department of Experimental and Health Sciences Pompeu Fabra University Carrer Doctor Aiguader 88 Barcelona 08003 Spain
| | - Paula Zamora‐Perez
- Integrative Biomedical Materials and Nanomedicine Lab Department of Experimental and Health Sciences Pompeu Fabra University Carrer Doctor Aiguader 88 Barcelona 08003 Spain
| | - Miriam Granado
- Nanomaterials for Bioimaging Group Departamento de Fisiología Facultad de Medicina Universidad Autónoma de Madrid C/ Arzobispo Morcillo s/n Madrid 28029 Spain
| | - Daniel González‐Hedström
- Nanomaterials for Bioimaging Group Departamento de Fisiología Facultad de Medicina Universidad Autónoma de Madrid C/ Arzobispo Morcillo s/n Madrid 28029 Spain
| | - María Fuente‐Fernández
- Nanomaterials for Bioimaging Group Departamento de Fisiología Facultad de Medicina Universidad Autónoma de Madrid C/ Arzobispo Morcillo s/n Madrid 28029 Spain
| | - Ángel Luis García‐Villalón
- Nanomaterials for Bioimaging Group Departamento de Fisiología Facultad de Medicina Universidad Autónoma de Madrid C/ Arzobispo Morcillo s/n Madrid 28029 Spain
| | - Laura Andrés‐Delgado
- Departamento de Anatomía Histología y Neurociencia Facultad de Medicina. Universidad Autónoma de Madrid. C/ Arzobispo Morcillo s/n Madrid 28029 Spain
| | - Emma Martín Rodríguez
- Instituto Ramón y Cajal de Investigación Sanitaria Hospital Ramón y Cajal Ctra. Colmenar km. 9,100 Madrid 28034 Spain
- Nanomaterials for Bioimaging Group Departamento de Física Aplicada Universidad Autónoma de Madrid C/ Francisco Tomás y Valiente 7 Madrid 28049 Spain
| | - Río Aguilar
- Cardiology Department Hospital Universitario de la Princesa Instituto Investigación Sanitaria Princesa (IIS‐IP) CIBER‐CV Universidad Autónoma de Madrid Calle Diego de León, 62 Madrid 28006 Spain
| | - Fernando Alfonso
- Cardiology Department Hospital Universitario de la Princesa Instituto Investigación Sanitaria Princesa (IIS‐IP) CIBER‐CV Universidad Autónoma de Madrid Calle Diego de León, 62 Madrid 28006 Spain
| | - José García Solé
- Nanomaterials for Bioimaging Group Departamento de Física de Materiales Universidad Autónoma de Madrid C/ Francisco Tomás y Valiente 7 Madrid 28049 Spain
| | - Pilar Rivera Gil
- Integrative Biomedical Materials and Nanomedicine Lab Department of Experimental and Health Sciences Pompeu Fabra University Carrer Doctor Aiguader 88 Barcelona 08003 Spain
| | - Daniel Jaque
- Nanomaterials for Bioimaging Group Departamento de Física de Materiales Universidad Autónoma de Madrid C/ Francisco Tomás y Valiente 7 Madrid 28049 Spain
- Instituto Ramón y Cajal de Investigación Sanitaria Hospital Ramón y Cajal Ctra. Colmenar km. 9,100 Madrid 28034 Spain
| | - Fernando Rivero
- Cardiology Department Hospital Universitario de la Princesa Instituto Investigación Sanitaria Princesa (IIS‐IP) CIBER‐CV Universidad Autónoma de Madrid Calle Diego de León, 62 Madrid 28006 Spain
| |
Collapse
|
7
|
Oligonucleotide-based Preconditioning of DCD Cardiac Donors and Its Impact on Cardiac Viability. Transplantation 2020; 103:2479-2485. [PMID: 31335774 DOI: 10.1097/tp.0000000000002849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND While clinical donation after circulatory death (DCD) cardiac transplantation is being implemented with increasing frequency to address the supply/demand mismatch of donor grafts, no research to date has examined a strategy of donor preconditioning to optimize the viability of DCD hearts for transplantation. In our rat model of the DCD protocol, we investigate the impact of pretreating donors with phosphorothioate-linked cytosine and guanine rich oligodeoxynucleotides (CpG ODN) and their effects on cardiac function, injury, and a novel left ventricular (LV) mRNA biomarker panel. METHODS DCD rats were subjected to a withdrawal protocol, followed by 20 minutes of warm acirculatory standoff, representing a group of severely injured hearts as previously demonstrated. Beating heart controls and DCD rats were pretreated with vehicle or stimulatory CpG ODN (beating heart control and DCD stimulated with CpG ODN, BST and DST). Hearts were harvested for ex situ heart perfusion (ESHP), where LV function, histochemical injury, and differences in gene expression were characterized between groups. RESULTS Donor pretreatment with CpG ODN doubled the number of functional DCD hearts at ESHP. Pretreatment was associated with improved systolic and diastolic LV function, a reduction in histological injury, and markedly reduced elaboration of cardiac troponin-I in coronary effluent during ESHP. Pretreatment was also associated with a reduction in mRNA biomarkers associated with myocardial injury. CONCLUSIONS A single dose of CpG ODN was associated with reduced biomarkers of cardiac injury and a 100% increase in cardiac viability in this rodent model of marginal DCD cardiac donation.
Collapse
|
8
|
Monocyte mimics improve mesenchymal stem cell-derived extracellular vesicle homing in a mouse MI/RI model. Biomaterials 2020; 255:120168. [PMID: 32562944 DOI: 10.1016/j.biomaterials.2020.120168] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/26/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022]
Abstract
Stem cell-derived extracellular vesicles (EVs) have been demonstrated to be effective in heart repair and regeneration post infarction. However, the poor homing efficiency and low yields of these therapeutics remain the major obstacles before they can be used in the clinic. To improve the delivery efficiency of EVs to ischemia-injured myocardium, we modified mesenchymal stem cell (MSC)-derived EVs with monocyte mimics through the method of membrane fusion. Monocyte mimic-bioinspired MSC-EVs (Mon-Exos) exhibited enhanced targeting efficiency to injured myocardium by mimicking the recruitment feature of monocytes after MI/RI, thus contributing to these exclusive adhesive molecules on monocyte mimics, particularly the Mac1/LFA1-ICAM-1 interaction. Through this strategy, Mon-Exos were shown to promote endothelial maturation during angiogenesis and modulate macrophage subpopulations after MI/RI, consistent with MSC-Exos biofunctions, and eventually improve therapeutic outcomes in cardiac function and pathohistology changes after treatments in a mouse MI/RI model. Ultimately, this strategy might provide us with a better way to assess the effects of stem cell EVs and offer additional techniques to help clinicians better manage regenerative therapeutics for ischemic heart diseases.
Collapse
|
9
|
Hofbauer TM, Mangold A, Scherz T, Seidl V, Panzenböck A, Ondracek AS, Müller J, Schneider M, Binder T, Hell L, Lang IM. Neutrophil extracellular traps and fibrocytes in ST-segment elevation myocardial infarction. Basic Res Cardiol 2019; 114:33. [PMID: 31312919 PMCID: PMC6647191 DOI: 10.1007/s00395-019-0740-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 06/26/2019] [Indexed: 01/22/2023]
Abstract
Leukocyte-mediated inflammation is central in atherothrombosis and ST-segment elevation myocardial infarction (STEMI). Neutrophil extracellular traps (NETs) have been shown to enhance atherothrombosis and stimulate fibroblast function. We analyzed the effects of NETs on cardiac remodeling after STEMI. We measured double-stranded (ds)DNA and citrullinated histone H3 (citH3) as NET surrogate markers in human culprit site and femoral blood collected during primary percutaneous coronary intervention (n = 50). Fibrocytes were characterized in whole blood by flow cytometry, and in culprit site thrombi and myocardium by immunofluorescence. To investigate mechanisms of fibrocyte activation, isolated NETs were used to induce fibrocyte responses in vitro. Enzymatic infarct size was assessed using creatine-phosphokinase isoform MB area under the curve. Left ventricular function was measured by transthoracic echocardiography. NET surrogate markers were increased at the culprit site compared to the femoral site and were positively correlated with infarct size and left ventricular dysfunction at follow-up. In vitro, NETs promoted fibrocyte differentiation from monocytes and induced fibrocyte activation. Highly activated fibrocytes accumulated at the culprit site and in the infarct transition zone. Our data suggest that NETs might be important mediators of fibrotic remodeling after STEMI, possibly by stimulating fibrocytes.
Collapse
Affiliation(s)
- Thomas M Hofbauer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Andreas Mangold
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Thomas Scherz
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Veronika Seidl
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Adelheid Panzenböck
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Anna S Ondracek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Julian Müller
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Matthias Schneider
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Thomas Binder
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Lena Hell
- Division of Haematology and Haemostaseology, Department of Internal Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Irene M Lang
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
10
|
Ibarra-Lara L, Sánchez-Aguilar M, Soria-Castro E, Vargas-Barrón J, Roldán FJ, Pavón N, Torres-Narváez JC, Cervantes-Pérez LG, Pastelín-Hernández G, Sánchez-Mendoza A. Clofibrate Treatment Decreases Inflammation and Reverses Myocardial Infarction-Induced Remodelation in a Rodent Experimental Model. Molecules 2019; 24:molecules24020270. [PMID: 30642049 PMCID: PMC6359129 DOI: 10.3390/molecules24020270] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 01/08/2023] Open
Abstract
Myocardial infarction (MI) initiates an inflammatory response that promotes both beneficial and deleterious effects. The early response helps the myocardium to remove damaged tissue; however, a prolonged later response brings cardiac remodeling characterized by functional, metabolic, and structural pathological changes. Current pharmacological treatments have failed to reverse ischemic-induced cardiac damage. Therefore, our aim was to study if clofibrate treatment was capable of decreasing inflammation and apoptosis, and reverse ventricular remodeling and MI-induced functional damage. Male Wistar rats were assigned to (1) Sham coronary artery ligation (Sham) or (2) Coronary artery ligation (MI). Seven days post-MI, animals were further divided to receive vehicle (V) or clofibrate (100 mg/kg, C) for 7 days. The expression of IL-6, TNF-α, and inflammatory related molecules ICAM-1, VCAM-1, MMP-2 and -9, nuclear NF-kB, and iNOS, were elevated in MI-V. These inflammatory biomarkers decreased in MI-C. Also, apoptotic proteins (Bax and pBad) were elevated in MI-V, while clofibrate augmented anti-apoptotic proteins (Bcl-2 and 14-3-3ε). Clofibrate also protected MI-induced changes in ultra-structure. The ex vivo evaluation of myocardial functioning showed that left ventricular pressure and mechanical work decreased in infarcted rats; clofibrate treatment raised those parameters to control values. Echocardiogram showed that clofibrate partially reduced LV dilation. In conclusion, clofibrate decreases cardiac remodeling, decreases inflammatory molecules, and partly preserves myocardial diameters.
Collapse
Affiliation(s)
- Luz Ibarra-Lara
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - María Sánchez-Aguilar
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - Elizabeth Soria-Castro
- Department of Pathology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - Jesús Vargas-Barrón
- Department of Haemodynamics, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - Francisco J Roldán
- Department of Haemodynamics, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - Natalia Pavón
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - Juan C Torres-Narváez
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - Luz G Cervantes-Pérez
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - Gustavo Pastelín-Hernández
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| | - Alicia Sánchez-Mendoza
- Department of Pharmacology, National Institute of Cardiology Ignacio Chávez, Juan Badiano No.1, Col. Sección XVI, Tlalpan, Z.C., Mexico City 14080, Mexico.
| |
Collapse
|
11
|
Abstract
Fundamental features of septic shock are vasodilation, increased permeability, hypovolemia, and ventricular dysfunction. Vasodilation owing to increased nitric oxide and prostaglandins is treated with vasopressors (norepinephrine first). Increased permeability relates to several pathways (Slit/Robo4, vascular endothelial growth factor, angiopoietin 1 and 2/Tie2 pathway, sphingosine-1-phosphate, and heparin-binding protein), some of which are targets for therapies. Hypovolemia is common and crystalloid is recommended for fluid resuscitation. Cardiomyocyte-inflammatory interactions decrease contractility and dobutamine is recommended to increase cardiac output. There is benefit in decreasing heart rate in selected patients with esmolol. Ivabradine is a novel agent for heart rate reduction without decreasing contractility.
Collapse
Affiliation(s)
- James A Russell
- Department of Medicine, Centre for Heart Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada.
| | - Barret Rush
- Division of Critical Care Medicine, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - John Boyd
- Department of Medicine, Centre for Heart Lung Innovation, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| |
Collapse
|
12
|
Hu P, Dai T, Yu W, Luo Y, Huang S. Intercellular adhesion molecule 1 rs5498 polymorphism is associated with the risk of myocardial infarction. Oncotarget 2017; 8:52594-52603. [PMID: 28881754 PMCID: PMC5581053 DOI: 10.18632/oncotarget.17529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/17/2017] [Indexed: 01/11/2023] Open
Abstract
Several studies addressed the association between Intercellular adhesion molecule 1 (ICAM-1) rs5498 polymorphism and Myocardial Infarction (MI) risk. However, they addressed conflicting findings. Therefore, the aim of this study was to explore whether ICAM-1 gene rs5498 polymorphism plays an important role in modifying the risk of MI. A meta-analysis was conducted on the association between ICAM-1 rs5498 polymorphism and MI. 12 eligible studies involving 1,696 cases and 3,039 controls were included in the meta-analysis. Meta-analysis revealed that ICAM-1 rs5498 polymorphism showed a strongly positive correlation with MI and could be viewed as a protective factor for MI. Furthermore, subgroup analysis according to ethnicity indicated that ICAM-1 rs5498 polymorphism decreased the risk of MI among Caucasian and Asian populations. In conclusion, ICAM-1 rs5498 polymorphism was associated with the decreased risk of MI. Larger sample size studies with more diverse ethnic populations are needed to confirm these findings.
Collapse
Affiliation(s)
- Pengfei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tao Dai
- Department of Cardiology, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Weiwei Yu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ying Luo
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shuwei Huang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Uehara Y, Murata Y, Shiga S, Hosoi Y. NSAIDs diclofenac, indomethacin, and meloxicam highly upregulate expression of ICAM-1 and COX-2 induced by X-irradiation in human endothelial cells. Biochem Biophys Res Commun 2016; 479:847-852. [PMID: 27687548 DOI: 10.1016/j.bbrc.2016.09.120] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 09/23/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND It is well known that radiation exposure to the heart and the use of non-steroidal anti-inflammatory drugs (NSAIDs) increase the risk of myocardial infarction (MI). Some NSAIDs are also known to act synergistically with ionizing radiation and have radio-sensitizing effects in radiotherapy. These evidences suggest that NSAIDs may affect the risk of MI after radiation exposure to the heart. In the present study, we investigated effects of NSAIDs on radiation-induced expression of cell adhesion molecules and COX-2, which are associated with inflammation and an increased risk of MI, in human endothelial cells. METHODS Effects of NSAIDs on radiation-induced expression of ICAM-1, VCAM-1, E-selectin, and COX-2 were investigated in human umbilical vein endothelial cells (HUVECs). As NSAIDs, diclofenac, etodolac, indomethacin, ketoprofen, meloxicam, and rofecoxib were used. RESULTS Irradiation with 10 Gy increased expression of ICAM-1 and COX-2, but it did not affect expression of VCAM-1 or E-selectin. All the NSAIDs upregulated radiation-induced expression of ICAM-1 and COX-2. The extent of upregulation varied depending on the types of NSAIDs. Indomethacin, diclofenac, and meloxicam highly upregulated radiation-induced expression of ICAM-1 and COX-2. The extent of upregulation was not related to the degree of COX-2 selectivity. An NF-κB inhibitor BAY 11-7082 suppressed radiation-induced expression of ICAM-1, but it did not suppress upregulated expression of ICAM-1 or COX-2 by combination treatment with X-irradiation and meloxicam, suggesting the existence of NF-κB-independent pathways for ICAM-1 and COX-2 induction. CONCLUSION Indomethacin, diclofenac, and meloxicam highly upregulated radiation-induced expression of ICAM-1 and COX-2 in HUVECs, which suggests that use of these NSAIDs may increase the effects of ionizing radiation and affect the risk of MI after radiation exposure to the heart.
Collapse
Affiliation(s)
- Yoshihiko Uehara
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi-ken 980-8575, Japan
| | - Yasuhiko Murata
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi-ken 980-8575, Japan
| | - Soichiro Shiga
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi-ken 980-8575, Japan
| | - Yoshio Hosoi
- Department of Radiation Biology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi-ken 980-8575, Japan.
| |
Collapse
|
14
|
Abstract
Recent research has identified promising targets for therapeutic interventions aimed at modulating the inflammatory response in sepsis. Herein, the authors describe mechanisms involved in the clearance of pathogen toxin from the circulation and potential interventions aimed at enhancing clearance mechanisms. The authors also describe advances in the understanding of the innate immune response as potential therapeutic targets. Finally, novel potential treatment strategies aimed at decreasing vascular leak are discussed.
Collapse
Affiliation(s)
- Peter Bentzer
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; Division of Critical Care Medicine, St. Paul's Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada; Department of Anesthesiology and Intensive Care, Lund University, Lund SE-221 85, Sweden
| | - James A Russell
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; Division of Critical Care Medicine, St. Paul's Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Keith R Walley
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; Division of Critical Care Medicine, St. Paul's Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada.
| |
Collapse
|
15
|
Meléndez GC, Manteufel EJ, Dehlin HM, Register TC, Levick SP. Non-human primate and rat cardiac fibroblasts show similar extracellular matrix-related and cellular adhesion gene responses to substance P. Heart Lung Circ 2015; 24:395-403. [PMID: 25550118 PMCID: PMC4492475 DOI: 10.1016/j.hlc.2014.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 11/12/2014] [Accepted: 11/19/2014] [Indexed: 11/25/2022]
Abstract
BACKGROUND The sensory nerve neuropeptide substance P (SP) regulates cardiac fibrosis in rodents under pressure overload conditions. Interestingly, SP induces transient increased expression of specific genes in isolated rat cardiac fibroblasts, without resultant changes in cell function. This suggests that SP 'primes' fibroblasts, but does not directly activate them. We investigated whether these unusual findings are specific to rodent fibroblasts or are translatable to a larger animal model more closely related to humans. METHODS We compared the effects of SP on genes associated with extracellular matrix (ECM) regulation, cell-cell adhesion, cell-matrix adhesion and ECM in cardiac fibroblasts isolated from a non-human primate and Sprague-Dawley rats. RESULTS We found that rodent and non-human primate cardiac fibroblasts showed similar responses in genes that relate to ECM regulation and cell adhesion in response to SP. There were large discrepancies in ECM component genes, however, this did not result in collagen or laminin synthesis in rat or non-human primate fibroblasts in response to SP. CONCLUSIONS This study further supports the notion that SP serves as a 'primer' for fibroblasts rather than initiating direct effects and suggests that rodent fibroblasts are a suitable model for studying gene and functional responses to SP in the absence of human or non-human primate fibroblasts.
Collapse
Affiliation(s)
- Giselle C Meléndez
- Department of Internal Medicine, Section on Cardiology, Wake Forest School of Medicine, Winston-Salem, NC; Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Edward J Manteufel
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI
| | - Heather M Dehlin
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI
| | - Thomas C Register
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Scott P Levick
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
16
|
Cohen MM. Perspectives on RAGE signaling and its role in cardiovascular disease. Am J Med Genet A 2013; 161A:2750-5. [PMID: 24123885 DOI: 10.1002/ajmg.a.36181] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 07/16/2013] [Indexed: 01/14/2023]
Abstract
RAGE stands for Receptor of Advanced Glycation Endproducts. The two main topics discussed are (1) the nature of RAGE signaling and (2) its role in cardiovascular disease. RAGE may occur in membrane-bound form or in secretory form. RAGE signaling involves multiple ligands: (1) several AGEs (2) amyloid β pecursor protein (APP), (3) high mobility group box 1 (HMGB1), (4) S100A4, (5) S100A8/A9, and (6) S100A12, which are calcium-binding proteins, and (7) S100B, a glial-derived protein. RAGE ligands and various diseases involving RAGE signaling are summarized in tabular form.
Collapse
Affiliation(s)
- M Michael Cohen
- Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
17
|
Behnes M, Ruff C, Lang S, Kälsch T, Borggrefe M, Elmas E. Intercellular adhesion molecule 1 (ICAM-1) - A new substrate for the development of ventricular fibrillation? Int J Cardiol 2013; 168:4917-9. [DOI: 10.1016/j.ijcard.2013.07.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 07/08/2013] [Indexed: 11/26/2022]
|
18
|
Zhang DW, Huang XZ, Wu JH, Fan YP, Shi H. Effects of Intercellular Adhesion Molecule-1 on Renal Damage in Spontaneously Hypertensive Rats. Ren Fail 2012; 34:915-20. [PMID: 22681549 DOI: 10.3109/0886022x.2012.692751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
19
|
Bidmon B, Kratochwill K, Rusai K, Kuster L, Herzog R, Eickelberg O, Aufricht C. Increased immunogenicity is an integral part of the heat shock response following renal ischemia. Cell Stress Chaperones 2012; 17:385-97. [PMID: 22180342 PMCID: PMC3312958 DOI: 10.1007/s12192-011-0314-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 11/29/2011] [Accepted: 11/30/2011] [Indexed: 11/25/2022] Open
Abstract
Renal ischemia increases tubular immunogenicity predisposing to increased risk of kidney allograft rejection. Ischemia-reperfusion not only disrupts cellular homeostasis but also induces the cytoprotective heat shock response that also plays a major role in cellular immune and defense processes. This study therefore tested the hypothesis that upregulation of renal tubular immunogenicity is an integral part of the heat shock response after renal ischemia. Expressions of 70 kDa heat shock protein (Hsp70), major histocompatibility complex (MHC) class II, and intercellular adhesion molecule-1 (ICAM-1) were assessed in normal rat kidney (NRK) cells following ATP depletion (antimycin A for 3 h) and heat (42°C for 24 h). In vitro, transient Hsp70 transfection and heat shock factor-1 (HSF-1) transcription factor decoy treatment were performed. In vivo, ischemic renal cortex was investigated in Sprague-Dawley rats following unilateral renal artery clamping for 45 min and 24 h recovery. Upregulation of Hsp70 was closely and significantly correlated with upregulation of MHC class II and/or ICAM-1 following ATP depletion and heat injury. Bioinformatics analysis searching the TRANSFAC database predicted HSF-1 binding sites in these genes. HSF-1 decoy significantly reduced the expression of immunogenicity markers in stressed NRK cells. In the in vivo rat model of renal ischemia, concordant upregulation of MHC class II molecules and Hsp70 suggests biological relevance of this link. The results demonstrate that upregulation of renal tubular immunogenicity is an integral part of the heat shock response after renal ischemia. Bioinformatic analysis predicted a molecular link to tubular immunogenicity at the level of the transcription factor HSF-1 that was experimentally verified by HSF-1 decoy treatment. Future studies in HSF-1 knockout mice are needed.
Collapse
Affiliation(s)
- Bettina Bidmon
- Department of Pediatrics, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Klaus Kratochwill
- Department of Pediatrics, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Krisztina Rusai
- Department of Pediatrics, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Lilian Kuster
- Department of Pediatrics, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Rebecca Herzog
- Department of Pediatrics, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, University Hospital, Helmholtz Zentrum München, University of Munich, Max-Lebsche-Platz 31, 81377 Munich, Germany
| | - Christoph Aufricht
- Department of Pediatrics, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| |
Collapse
|
20
|
Marchant DJ, Boyd JH, Lin DC, Granville DJ, Garmaroudi FS, McManus BM. Inflammation in myocardial diseases. Circ Res 2012; 110:126-44. [PMID: 22223210 DOI: 10.1161/circresaha.111.243170] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammatory processes underlie a broad spectrum of conditions that injure the heart muscle and cause both structural and functional deficits. In this article, we address current knowledge regarding 4 common forms of myocardial inflammation: myocardial ischemia and reperfusion, sepsis, viral myocarditis, and immune rejection. Each of these pathological states has its own unique features in pathogenesis and disease evolution, but all reflect inflammatory mechanisms that are partially shared. From the point of injury to the mobilization of innate and adaptive immune responses and inflammatory amplification, the cellular and soluble mediators and mechanisms examined in this review will be discussed with a view that both beneficial and adverse consequences arise in these human conditions.
Collapse
Affiliation(s)
- David J Marchant
- James Hogg Research Centre and Institute for Heart + Lung Health, Department of Pathology and Laboratory Medicine, University of British Columbia, Providence Health Care, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
21
|
Turner NA, Das A, O’Regan DJ, Ball SG, Porter KE. Human cardiac fibroblasts express ICAM-1, E-selectin and CXC chemokines in response to proinflammatory cytokine stimulation. Int J Biochem Cell Biol 2011; 43:1450-8. [PMID: 21718796 DOI: 10.1016/j.biocel.2011.06.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/13/2011] [Accepted: 06/14/2011] [Indexed: 11/17/2022]
|
22
|
Mathur S, Walley KR, Wang Y, Indrambarya T, Boyd JH. Extracellular heat shock protein 70 induces cardiomyocyte inflammation and contractile dysfunction via TLR2. Circ J 2011; 75:2445-52. [PMID: 21817814 DOI: 10.1253/circj.cj-11-0194] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Toll-like receptors (TLRs) are expressed on cardiomyocytes and recognize pathogen-associated molecular patterns. Whether endogenous molecules produced by tissue injury (damage associated molecular patterns, DAMPs) can induce cardiomyocyte inflammation via TLR signalling pathways and/or reduce cardiomyocyte contractility is unknown. METHODS AND RESULTS Primary cardiomyocytes isolated from nuclear factor κ B (NFκB)-luciferase knock-in mice were used to assess NFκB signalling. DAMPs, HSP60, HSP70 and HMGB1, increased NFκB transcriptional activity compared to controls. HSP70 stood out compared to other DAMPs and even lipopolysaccharide (LPS). Subsequent experiments focused on HSP70. Cardiomyocytes exposed to HSP70 had a 58% decrease in contractility without a decrease in calcium flux. Exposure of cultured HL-1 cardiomyocytes to HSP70 resulted in increased expression of intercellular adhesion molecule 1 (ICAM-1), interleukin 6 (IL-6) and keratinocyte-derived chemokine (KC) compared to controls. Knock-out mice for TLR2, TLR4 and MyD88, plus background strain controls (C57BL/6) were used to assess induction of cardiomyocyte inflammation by HSP70. The cardiomyocyte expression of ICAM-1 induced by HSP70 was significantly reduced in TLR2 and MyD88 knock-out mice but not TLR4 knock-out mice; implicating the TLR2 signalling pathway. Furthermore, blocking antibodies to TLR2 were able to abrogate HSP70-induced contractile dysfunction and cell death. CONCLUSIONS Extracellular HSP70 acting via TLR2 and its obligate downstream adaptor molecule, MyD88, activate NFκB. This causes cardiomyocyte inflammation and decreased contractility.
Collapse
Affiliation(s)
- Sumeet Mathur
- Critical Care Research Laboratories, Heart+Lung Institute at St. Paul's Hospital, University of British Columbia, Canada
| | | | | | | | | |
Collapse
|
23
|
Suzuki JI, Ogawa M, Takayama K, Taniyama Y, Morishita R, Hirata Y, Nagai R, Isobe M. Ultrasound-Microbubble–Mediated Intercellular Adhesion Molecule-1 Small Interfering Ribonucleic Acid Transfection Attenuates Neointimal Formation After Arterial Injury in Mice. J Am Coll Cardiol 2010; 55:904-13. [DOI: 10.1016/j.jacc.2009.09.054] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 09/03/2009] [Accepted: 09/15/2009] [Indexed: 01/19/2023]
|
24
|
Boyd JH, Kan B, Roberts H, Wang Y, Walley KR. S100A8 and S100A9 mediate endotoxin-induced cardiomyocyte dysfunction via the receptor for advanced glycation end products. Circ Res 2008; 102:1239-46. [PMID: 18403730 DOI: 10.1161/circresaha.107.167544] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cardiovascular dysfunction as a result of sepsis is the leading cause of death in the critically ill. Cardiomyocytes respond to infectious pathogens with a Toll-like receptor-initiated proinflammatory response in conjunction with a decrease in contractility, although the downstream events linking Toll-like receptor activation and reduced cardiac contractility remain to be elucidated. Using microarray analysis of cardiac tissue exposed to systemic lipopolysaccharide (LPS), we discovered that 2 small calcium-regulating proteins (S100A8 and S100A9) are highly upregulated. HL-1 cardiomyocytes, isolated primary cardiomyocytes, and live mice were exposed to LPS, whereas beating HL-1 cells had S100A8 and S100A9 overexpressed and their calcium flux quantified. Using in vivo microbubble technology, we delivered S100A8 and S100A9 to normal mouse hearts; using the same technology, we inhibited S100A9 production in mouse hearts and subsequently exposed them to LPS. Coimmunoprecipitation of S100A8 and S100A9 identified interaction with RAGE (the receptor for advanced glycation end products), the cardiac function and postreceptor signaling of which were investigated. HL-1 cardiomyocytes, isolated primary cardiomyocytes, and whole hearts exposed to LPS have large increases in S100A8 and S100A9. Cardiac overexpression of S100A8 and S100A9 led to a RAGE-dependent decrease in calcium flux and, in the intact mouse, to a decreased cardiac ejection fraction, whereas knockdown of S100A9 attenuated LPS-induced cardiac dysfunction. Cardiomyocytes exposed to LPS express S100A8 and S100A9, leading to a RAGE-mediated decrease in cardiomyocyte contractility. This finding provides a novel mechanistic link between circulating pathogen-associated molecular products and subsequent cardiac dysfunction.
Collapse
Affiliation(s)
- John H Boyd
- Critical Care Research Laboratories, St. Paul' Hospital, University of British Columbia, Vancouver, Canada.
| | | | | | | | | |
Collapse
|
25
|
Sodium/hydrogen exchange inhibition with cariporide reduces leukocyte adhesion via P-selectin suppression during inflammation. Br J Pharmacol 2008; 153:1678-85. [PMID: 18332863 DOI: 10.1038/sj.bjp.0707647] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE The Na(+)/H(+) exchange (NHE) inhibitor cariporide is known to ameliorate ischaemia/reperfusion (I/R) injury by reduction of cytosolic Ca(2+) overload. Leukocyte activation and infiltration also mediates I/R injury but whether cariporide reduces I/R injury by affecting leukocyte activation is unknown. We studied the effect of cariporide on thrombin and I/R induced leukocyte activation and infiltration models and examined P-selectin expression as a potential mechanism for any identified effects. EXPERIMENTAL APPROACH An in vivo rat mesenteric microcirculation microscopy model was used with stimulation by thrombin (0.5 micro ml(-1)) superfusion or ischaemia (by haemorrhagic shock for 60 min) and reperfusion (90 min). KEY RESULTS Treatment with cariporide (10 mg kg(-1) i.v.) significantly reduced leukocyte rolling, adhesion and extravasation after thrombin exposure. Similarly, cariporide reduced leukocyte rolling (54+/-6.2 to 2.4+/-1.0 cells min(-1), P<0.01), adherence (6.3+/-1.9 to 1.2+/-0.4 cells 100 microm(-1), P<0.01) and extravasation (9.1+/-2.1 to 2.4+/-1.1 cells per 20 x 100 microm perivascular space, P<0.05), following haemorrhagic shock induced systemic ischaemia and reperfusion. The cell adhesion molecule P-selectin showed a profound decrease in endothelial expression following cariporide administration in both thrombin and I/R stimulated groups (35.4+/-3.2 vs 14.2+/-4.1% P-selectin positive cells per tissue section, P<0.01). CONCLUSIONS AND IMPLICATIONS The NHE inhibitor cariporide is known to limit reperfusion injury by controlling Ca(2+) overload but these data are novel evidence for a vasculoprotective effect of NHE inhibition at all levels of leukocyte activation, an effect which is likely to be mediated at least in part by a reduction of P-selectin expression.
Collapse
|
26
|
Tziakas DN, Chalikias GK, Kaski JC, Kekes A, Hatzinikolaou EI, Stakos DA, Tentes IK, Kortsaris AX, Hatseras DI. Inflammatory and anti-inflammatory variable clusters and risk prediction in acute coronary syndrome patients: A factor analysis approach. Atherosclerosis 2007; 193:196-203. [PMID: 16857204 DOI: 10.1016/j.atherosclerosis.2006.06.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 06/04/2006] [Accepted: 06/14/2006] [Indexed: 11/15/2022]
Abstract
BACKGROUND Numerous inflammatory mediators such as C-reactive protein (CRP), fibrinogen, interleukin-18 (IL-18), and inter-cellular adhesion molecule-1 (ICAM-1) have been proposed for risk stratification in acute coronary syndrome (ACS) patients. However, interactions between these markers have made it difficult to assess their true role in risk prediction. Factor analysis is a multivariable statistical technique that reduces a large number of intercorrelated variables to a smaller set of independent clusters, underlining physiological relationships. The aim of this study was to investigate, using factor analysis, a clustering of pro-inflammatory markers, anti-inflammatory cytokines such as interleukin-10 (IL-10) and HDL cholesterol, and to determine their role in prediction of risk of recurrent coronary events in ACS patients. METHODS We assessed 320 consecutive patients (236 men; 67 years; IQ 58-74 years) admitted with ACS. The composite of cardiac death and re-hospitalization with non-fatal myocardial infarction, or unstable angina, was the pre-specified study end-point. Serum CRP, fibrinogen, HDL cholesterol, IL-10, IL-18 and ICAM-1 levels were measured at study entry. We assessed independent predictors of the combined end-point during a 1-year follow-up using multiple logistic regression analysis. RESULTS Factor analysis identified three clusters which were arbitrarily interpreted as (1) a "systemic inflammation" cluster with positive loadings of CRP and fibrinogen, (2) a "local inflammation-endothelial dysfunction" cluster with positive loadings of IL-18 and ICAM-1 and (3) an "anti-inflammation" cluster comprising IL-10 and HDL cholesterol. Only the "anti-inflammation" cluster was a significant predictor (OR 0.66, 95% CI: 0.49-0.89) of adverse cardiac events during a 1-year follow-up and remained significant (OR 0.65, 95% CI: 0.48-0.88) in a multivariate model that included all three factors. CONCLUSIONS Although inflammatory markers such as CRP predict future cardiovascular events in ACS patients, when all inflammatory mediators are taken into account in a prospective analysis of risk, markers reflecting anti-inflammatory mechanisms are better prognostic markers.
Collapse
Affiliation(s)
- Dimitrios N Tziakas
- University Cardiology Department, Democritus University of Thrace, Voulgaroktonou 23, 68100 Alexandroupolis, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Schramm R, Menger MD, Kirsch S, Langer F, Harder Y, Hamacher J, Schäfers HJ. The subepicardial microcirculation in heterotopically transplanted mouse hearts: An intravital multifluorescence microscopy study. J Thorac Cardiovasc Surg 2007; 134:210-7, 217.e1. [PMID: 17599511 DOI: 10.1016/j.jtcvs.2007.02.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 01/09/2007] [Accepted: 02/12/2007] [Indexed: 12/17/2022]
Abstract
OBJECTIVE We developed a model for intravital microscopic analysis of the coronary microcirculation in transplanted murine hearts and assessed the influence of cold ischemia on postischemic microcirculatory dysfunctions. METHODS Murine hearts were exposed to 60 (n = 12) and 240 minutes (n = 8) of cold ischemia before syngeneic heterotopic transplantation. Intravital fluorescence microscopy allowed detailed analysis of the right ventricular coronary microcirculation, including feeding arterioles, nutritive capillaries, and postcapillary venules. With this technique, we further studied leukocyte-endothelial cell interactions, microvascular permeability, tissue oxygenation, and microlymphatics. RESULTS Cold ischemia of 240 minutes aggravated nutritive capillary perfusion failure, indicated by a significant reduction of functional capillary density and capillary flow velocity by 63% and 45% (P < .05 vs 60-minute cold ischemic isografts). The mitochondrial redox state, visualized by nicotinamide adenine dinucleotide hydrogen autofluorescence, was markedly deteriorated after 240-minute cold ischemia (P < .05), indicating a persistent mismatch between oxygen supply and demand resulting from pronounced capillary no-reflow. Prolonged ischemia further resulted in 6- and 11-fold higher numbers of rolling and firmly adherent leukocytes in postcapillary venules (P < .05), together with increased microvascular permeability. CONCLUSIONS We introduce a novel approach to visualize in detail the murine coronary microcirculation in vivo by multifluorescence microscopy. Our data demonstrate that prolonged cold ischemia provokes posttransplant capillary no-reflow, leukocytic inflammation, and persistent tissue hypoxia.
Collapse
Affiliation(s)
- René Schramm
- Department of Thoracic and Cardiovascular Surgery, University of Saarland, Homburg/Saar, Germany.
| | | | | | | | | | | | | |
Collapse
|
28
|
Sapna S, Shivakumar K. Hypoxia and antioxidants enhance soluble ICAM-1 release from cardiac fibroblasts. Mol Cell Biochem 2007; 303:259-62. [PMID: 17458516 DOI: 10.1007/s11010-007-9475-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Accepted: 04/03/2007] [Indexed: 11/29/2022]
Abstract
Intercellular adhesion molecule-1 plays a key role in mediating inflammatory and immune responses. There is also increasing appreciation of the role of its soluble form, sICAM-1, in regulating inflammation. This study evaluated the effects of hypoxia and N-acetyl-L-cysteine on sICAM-1 production by adult rat cardiac fibroblasts. By ELISA, hypoxia was found to cause a 61% increase in sICAM-1 in cardiac fibroblast culture supernates. However, RT-PCR did not reveal a concomitant increase in cell surface ICAM-1 transcript levels, suggesting that the increase in sICAM-1 may involve post-transcriptional and/or post-translational mechanisms. Using pharmacological inhibitors, it was observed that p42/44 MAPK and PKC mediate the stimulatory effect of hypoxia on sICAM-1 production. Remarkably, N-acetyl-L-cysteine caused a 3-fold increase in sICAM-1 by p42/44 MAPK-, p38 MAPK- and PKC-independent mechanisms. Pyrrolidine dithiocarbamate, another potent antioxidant, also augmented sICAM-1. The findings presented in this communication underscore the link between redox status and sICAM-1 release from cardiac fibroblasts. Further, because hypoxia is a major component of myocardial ischemia and is pro-inflammatory, and both N-acetylcysteine and pyrrolidine dithiocarbamate are clinically used antioxidants, the observations may have clinical significance.
Collapse
Affiliation(s)
- S Sapna
- Division of Cellular and Molecular Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, 695 011, Kerala, India
| | | |
Collapse
|
29
|
Kaatz M, Berod L, Lagadari M, Fluhr JW, Elsner P, Norgauer J. Microtubules Regulate Expression of ICAM-1 in Epidermoid Cells (KB Cells). Skin Pharmacol Physiol 2006; 19:322-8. [PMID: 16931899 DOI: 10.1159/000095252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2005] [Accepted: 01/30/2006] [Indexed: 11/19/2022]
Abstract
The intercellular adhesion molecule-1/CD54 (ICAM-1) functions as a counterreceptor for other adhesion molecules (e.g. the lymphocyte function-associated antigen-1/CD11a/CD18) required for the interaction of a large variety of cells with leucocytes. Constitutive expression of ICAM-1 in human epidermoid cells (KB cells) is low, but inducible by interferon-gamma (IFN-gamma). Treatment of KB cells with microtubule-disrupting agents, like colchicine, nocodazole and vinblastine, potentiated the constitutive and cytokine-induced ICAM-1 expression on the cell surface. Actinomycin D inhibited microtubule-disrupting agent-induced ICAM-1 surface expression. Increased steady-state levels of ICAM-1 transcripts were found after treatment of KB cells with microtubule-disrupting agents. However, microtubule-disrupting agents neither altered the glyceraldehyde-3-phosphate dehydrogenase mRNA levels nor the amount of expressed alpha(2)-, alpha(3)-and beta(1)-integrins at the cell surface. In addition, they did not change the ICAM-1 mRNA half-life. These studies indicate a control function of the microtubule network on the expression of ICAM-1.
Collapse
Affiliation(s)
- M Kaatz
- Department of Dermatology, University of Jena, Jena, Germany
| | | | | | | | | | | |
Collapse
|
30
|
Nahrendorf M, Hu K, Frantz S, Jaffer FA, Tung CH, Hiller KH, Voll S, Nordbeck P, Sosnovik D, Gattenlöhner S, Novikov M, Dickneite G, Reed GL, Jakob P, Rosenzweig A, Bauer WR, Weissleder R, Ertl G. Factor XIII deficiency causes cardiac rupture, impairs wound healing, and aggravates cardiac remodeling in mice with myocardial infarction. Circulation 2006; 113:1196-202. [PMID: 16505171 PMCID: PMC4066325 DOI: 10.1161/circulationaha.105.602094] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Identification of key molecular players in myocardial healing could lead to improved therapies, reduction of scar formation, and heart failure after myocardial infarction (MI). We hypothesized that clotting factor XIII (FXIII), a transglutaminase involved in wound healing, may play an important role in MI given prior clinical and mouse model data. METHODS AND RESULTS To determine whether a truly causative relationship existed between FXIII activity and myocardial healing, we prospectively studied myocardial repair in FXIII-deficient mice. All FXIII(-/-) and FXIII(-)(/+) (FXIII activity <5% and 70%) mice died within 5 days after MI from left ventricular rupture. In contradistinction, FXIII(-/-) mice that received 5 days of intravenous FXIII replacement therapy had normal survival rates; however, cardiac MRI demonstrated worse left ventricular remodeling in these reconstituted FXIII(-/-) mice. Using a FXIII-sensitive molecular imaging agent, we found significantly greater FXIII activity in wild-type mice and FXIII(-/-) mice receiving supplemental FXIII than in FXIII(-/-) mice (P<0.05). In FXIII(-/-) but not in reconstituted FXIII(-/-) mice, histology revealed diminished neutrophil migration into the MI. Reverse transcriptase-polymerase chain reaction studies suggested that the impaired inflammatory response in FXIII(-/-) mice was independent of intercellular adhesion molecule and lipopolysaccharide-induced CXC chemokine, both important for cell migration. After MI, expression of matrix metalloproteinase-9 was 650% higher and collagen-1 was 53% lower in FXIII(-/-) mice, establishing an imbalance in extracellular matrix turnover and providing a possible mechanism for the observed cardiac rupture in the FXIII(-/-) mice. CONCLUSIONS These data suggest that FXIII has an important role in murine myocardial healing after infarction.
Collapse
Affiliation(s)
- Matthias Nahrendorf
- Medizinische Klinik und Poliklinik I, Universität Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Affiliation(s)
- Ehrin J Armstrong
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | |
Collapse
|
32
|
Herron BJ, Rao C, Liu S, Laprade L, Richardson JA, Olivieri E, Semsarian C, Millar SE, Stubbs L, Beier DR. A mutation in NFkB interacting protein 1 results in cardiomyopathy and abnormal skin development in wa3 mice. Hum Mol Genet 2005; 14:667-77. [PMID: 15661756 DOI: 10.1093/hmg/ddi063] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
We have identified waved 3 (wa3), a novel recessive mutation that causes abnormalities of the heart and skin. The cardiac defect results in a severe and rapidly progressive dilated cardiomyopathy. We identified the gene mutated in these mice, which we call NFkB interacting protein1 (Nkip1), using positional cloning. Nkip1 is expressed in skin, heart and vascular endothelium and shares homology with a small family of proteins that play a role in the regulation of transcription factors. A C-terminal fragment of this protein was previously identified as the RelA associated inhibitor (RAI). We show that the full-length protein is larger than previously described, and we confirm that it interacts with NFkB in vivo. Expression analysis of genes known to be regulated by NFkB revealed that Intercellular adhesion molecule 1 (Icam1) expression is consistently elevated in mutant mice. This result suggests that wa3 mutant mice represent a potentially important model for the analysis of the role of inflammatory processes in heart disease.
Collapse
Affiliation(s)
- Bruce J Herron
- Genetics Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
|