1
|
Manji A, Wang L, Pape CM, McCaig LA, Troitskaya A, Batnyam O, McDonald LJ, Appleton CT, Veldhuizen RA, Gill SE. Effect of aging on pulmonary cellular responses during mechanical ventilation. JCI Insight 2025; 10:e185834. [PMID: 39946196 PMCID: PMC11949020 DOI: 10.1172/jci.insight.185834] [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: 08/13/2024] [Accepted: 01/28/2025] [Indexed: 03/25/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) results in substantial morbidity and mortality, especially in elderly people. Mechanical ventilation, a common supportive treatment for ARDS, is necessary for maintaining gas exchange but can also propagate injury. We hypothesized that aging leads to alterations in surfactant function, inflammatory signaling, and microvascular permeability within the lung during mechanical ventilation. Young and aged male mice were mechanically ventilated, and surfactant function, inflammation, and vascular permeability were assessed. Additionally, single-cell RNA-Seq was used to delineate cell-specific transcriptional changes. The results showed that, in aged mice, surfactant dysfunction and vascular permeability were significantly augmented, while inflammation was less pronounced. Differential gene expression and pathway analyses revealed that alveolar macrophages in aged mice showed a blunted inflammatory response, while aged endothelial cells exhibited altered cell-cell junction formation. In vitro functional analysis revealed that aged endothelial cells had an impaired ability to form a barrier. These results highlight the complex interplay between aging and mechanical ventilation, including an age-related predisposition to endothelial barrier dysfunction, due to altered cell-cell junction formation, and decreased inflammation, potentially due to immune exhaustion. It is concluded that age-related vascular changes may underlie the increased susceptibility to injury during mechanical ventilation in elderly patients.
Collapse
Affiliation(s)
- Aminmohamed Manji
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
| | - Lefeng Wang
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Cynthia M. Pape
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Lynda A. McCaig
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Alexandra Troitskaya
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
| | - Onon Batnyam
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
| | - Leah J.J. McDonald
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | | | - Ruud A.W. Veldhuizen
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
- Department of Medicine, and
| | - Sean E. Gill
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
- Department of Medicine, and
| |
Collapse
|
2
|
Lalitha AV, Vasudevan A, Moorthy M, Ramaswamy G. Profiling Molecular Changes of Host Response to Predict Outcome in Children with Septic Shock. Indian J Crit Care Med 2024; 28:879-886. [PMID: 39360202 PMCID: PMC11443272 DOI: 10.5005/jp-journals-10071-24789] [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: 04/13/2024] [Accepted: 07/23/2024] [Indexed: 10/04/2024] Open
Abstract
Background Septic shock is associated with high mortality and there is significant heterogeneity in the host response. The aim of this study was to understand the genome-wide expression transcriptomic signatures in children with septic shock and correlate them with outcomes. Methods This was a prospective study conducted on children (aged 1 month to 18 years) admitted to the PICU (June-December 2021) with septic shock. Demographic details, clinical details, and administered treatment were collected. Differential gene expression analysis was performed to understand the genes and pathways affecting in different subjects. Results Fifteen patients were recruited (Septic shock survivors (n = 5), nonsurvivors (n = 5), and non-sepsis controls (n = 5). The median age of the patients in survivors and nonsurvivors was 15 (13, 24) months and 180 (180, 184) months, respectively. The sepsis-survivors vs nonsepsis possessed 983 upregulated and 624 downregulated genes while comparing sepsis nonsurvivors (SNS) with nonsepsis yielded 1,854 upregulated and 1,761 downregulated genes. Further, the lowest number of deregulated genes (383 upregulated and 486 downregulated) were present in SNS compared to sepsis survivors. The major Reactome pathways, found upregulated in SNSs relative to survivors included CD22 mediated B cell receptor (BCR) regulation, scavenging of heme from plasma, and creation of C4 and C2 activators while T cell receptor (TCR) signaling, the common pathway of fibrin clot formation and generation of second messenger molecules were found to be downregulated. Conclusion Mortality-related gene signatures are promising diagnostic biomarkers for pediatric sepsis. How to cite this article Lalitha AV, Vasudevan A, Moorthy M, Ramaswamy G. Profiling Molecular Changes of Host Response to Predict Outcome in Children with Septic Shock. Indian J Crit Care Med 2024;28(9):879-886.
Collapse
Affiliation(s)
- A V Lalitha
- Department of Pediatric Critical Care, St. John's Medical College and Hospital, Bengaluru, Karnataka, India
| | - Anil Vasudevan
- Department of Pediatric Nephrology, St. John's Medical College and Hospital, Bengaluru, Karnataka, India
| | - Manju Moorthy
- Department of Research and Development, Theracues Innovations Pvt. Ltd., Bengaluru, Karnataka, India
| | - Gopalakrishna Ramaswamy
- Department of Research and Development, Theracues Innovations Pvt. Ltd., Bengaluru, Karnataka, India
| |
Collapse
|
3
|
Hu M, Meganathan I, Zhu J, MacArthur R, Kassiri Z. Loss of TIMP3, but not TIMP4, exacerbates thoracic and abdominal aortic aneurysm. J Mol Cell Cardiol 2023; 184:61-74. [PMID: 37844423 DOI: 10.1016/j.yjmcc.2023.10.001] [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: 02/13/2023] [Revised: 10/01/2023] [Accepted: 10/01/2023] [Indexed: 10/18/2023]
Abstract
AIMS Aorta exhibits regional heterogeneity (structural and functional), while different etiologies for thoracic and abdominal aortic aneurysm (TAA, AAA) are recognized. Tissue inhibitor of metalloproteinases (TIMPs) regulate vascular remodeling through different mechanisms. Region-dependent functions have been reported for TIMP3 and TIMP4 in vascular pathologies. We investigated the region-specific function of these TIMPs in development of TAA versus AAA. METHODS & RESULTS TAA or AAA was induced in male and female mice lacking TIMP3 (Timp3-/-), TIMP4 (Timp4-/-) or in wildtype (WT) mice by peri-adventitial elastase application. Loss of TIMP3 exacerbated TAA and AAA severity in males and females, with a greater increase in proteinase activity, smooth muscle cell phenotypic switching post-AAA and -TAA, while increased inflammation was detected in the media post-AAA, but in the adventitia post-TAA. Timp3-/- mice showed impaired intimal barrier integrity post-AAA, but a greater adventitial vasa-vasorum branching post-TAA, which could explain the site of inflammation in AAA versus TAA. Severity of TAA and AAA in Timp4-/- mice was similar to WT mice. In vitro, Timp3 knockdown more severely compromised the permeability of human aortic EC monolayer compared to Timp4 knockdown or the control group. In aneurysmal aorta specimens from patients, TIMP3 expression decreased in the media in AAA, and in adventitial in TAA specimens, consistent with the impact of its loss in AAA versus TAA in mice. CONCLUSION TIMP3 loss exacerbates inflammation, adverse remodeling and aortic dilation, but triggers different patterns of remodeling in AAA versus TAA, and through different mechanisms.
Collapse
Affiliation(s)
- Mei Hu
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ilamaran Meganathan
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jiechun Zhu
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rodrick MacArthur
- Department of Cardiac surgery, Mazankowski Alberta Heart Institute, University of Alberta Hospital, Edmonton, AB, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Manríquez-Treviño Y, Sánchez-Ramírez B, Grado-Ahuir JA, Castro-Valenzuela B, González-Horta C, Burrola-Barraza M. Human TIMP1 Is a Growth Factor That Improves Oocyte Developmental Competence. BIOTECH 2023; 12:60. [PMID: 37873882 PMCID: PMC10594479 DOI: 10.3390/biotech12040060] [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: 07/07/2023] [Revised: 08/24/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023] Open
Abstract
Oocyte developmental competence is the ability of a mature oocyte to be fertilized and subsequently support embryonic development. Such competence is gained during folliculogenesis and is facilitated by the bidirectional communication into a compacted cumulus-oocyte complex (COC). Human tissue inhibitor of metalloproteinases-1 (TIMP1) participates in biological processes, including cell growth, differentiation, and apoptosis. This study aimed to evaluate the influence of TIMP1 as a growth factor on the in vitro maturation (IVM) culture of bovine COCs to improve oocyte developmental competence. All TIMP1 treatments (50, 100, and 150 ng/mL) favored the COCs' compaction structure (p < 0.05). TIMP1 at 150 ng/mL produced more oocytes in metaphase II compared to the other treatments (p < 0.05). The 150 ng/mL TIMP1 generated oocytes with the most (p < 0.05) cortical granules below the plasma membrane (pattern I). In a parthenogenesis assay, oocyte IVM in 50 ng/mL of TIMP1 produced the most blastocyst compared to the other treatments (p < 0.05). The Principal Component Analysis (PCA) showed that 50 ng/mL of TIMP1 was the best condition to develop oocyte competence because it was associated with the COC compact and cortical granule pattern I. TIMP1 influences the development of oocyte competence when added to the IVM culture medium of COCs.
Collapse
Affiliation(s)
- Yolanda Manríquez-Treviño
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Blanca Sánchez-Ramírez
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua (UACH), Campus Universitario #2, Chihuahua 31125, Chihuahua, Mexico; (B.S.-R.); (C.G.-H.)
| | - Juan Alberto Grado-Ahuir
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Beatriz Castro-Valenzuela
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| | - Carmen González-Horta
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua (UACH), Campus Universitario #2, Chihuahua 31125, Chihuahua, Mexico; (B.S.-R.); (C.G.-H.)
| | - M.Eduviges Burrola-Barraza
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua (UACH), Perif. Fco. R. Almada Km. 1, Chihuahua 31453, Chihuahua, Mexico; (Y.M.-T.); (J.A.G.-A.); (B.C.-V.)
| |
Collapse
|
5
|
Liu Y, Ji X, Zhou Z, Zhang J, Zhang J. Myocardial ischemia-reperfusion injury; Molecular mechanisms and prevention. Microvasc Res 2023:104565. [PMID: 37307911 DOI: 10.1016/j.mvr.2023.104565] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
Cardiovascular diseases are one of the leading causes of mortality in developed countries. Among cardiovascular disorders, myocardial infarction remains a life-threatening problem predisposing to the development and progression of ischemic heart failure. Ischemia/reperfusion (I/R) injury is a critical cause of myocardial injury. In recent decades, many efforts have been made to find the molecular and cellular mechanisms underlying the development of myocardial I/R injury and post-ischemic remodeling. Some of these mechanisms are mitochondrial dysfunction, metabolic alterations, inflammation, high production of ROS, and autophagy deregulation. Despite continuous efforts, myocardial I/R injury remains a major challenge in medical treatments of thrombolytic therapy, heart disease, primary percutaneous coronary intervention, and coronary arterial bypass grafting. The development of effective therapeutic strategies to reduce or prevent myocardial I/R injury is of great clinical significance.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Xiang Ji
- Department of Integrative, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Zhou Zhou
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Jingwen Zhang
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Juan Zhang
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China; First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250011, China.
| |
Collapse
|
6
|
Jayawardena DP, Masciantonio MG, Wang L, Mehta S, DeGurse N, Pape C, Gill SE. Imbalance of Pulmonary Microvascular Endothelial Cell-Expression of Metalloproteinases and Their Endogenous Inhibitors Promotes Septic Barrier Dysfunction. Int J Mol Sci 2023; 24:ijms24097875. [PMID: 37175585 PMCID: PMC10178398 DOI: 10.3390/ijms24097875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023] Open
Abstract
Sepsis is a life-threatening disease characterized by excessive inflammation leading to organ dysfunction. During sepsis, pulmonary microvascular endothelial cells (PMVEC) lose barrier function associated with inter-PMVEC junction disruption. Matrix metalloproteinases (MMP) and a disintegrin and metalloproteinases (ADAM), which are regulated by tissue inhibitors of metalloproteinases (TIMPs), can cleave cell-cell junctional proteins, suggesting a role in PMVEC barrier dysfunction. We hypothesize that septic PMVEC barrier dysfunction is due to a disruption in the balance between PMVEC-specific metalloproteinases and TIMPs leading to increased metalloproteinase activity. The effects of sepsis on TIMPs and metalloproteinases were assessed ex vivo in PMVEC from healthy (sham) and septic (cecal ligation and perforation) mice, as well as in vitro in isolated PMVEC stimulated with cytomix, lipopolysaccharide (LPS), and cytomix + LPS vs. PBS. PMVEC had high basal Timp expression and lower metalloproteinase expression, and septic stimulation shifted expression in favour of metalloproteinases. Septic stimulation increased MMP13 and ADAM17 activity associated with a loss of inter-PMVEC junctional proteins and barrier dysfunction, which was rescued by treatment with metalloproteinase inhibitors. Collectively, our studies support a role for metalloproteinase-TIMP imbalance in septic PMVEC barrier dysfunction, and suggest that inhibition of specific metalloproteinases may be a therapeutic avenue for septic patients.
Collapse
Affiliation(s)
- Devika P Jayawardena
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Marcello G Masciantonio
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Lefeng Wang
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Division of Respirology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Sanjay Mehta
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Division of Respirology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Natalie DeGurse
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Cynthia Pape
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Division of Respirology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
7
|
Liao S, Lin Y, Liu L, Yang S, Lin Y, He J, Shao Y. ADAM10-a "multitasker" in sepsis: focus on its posttranslational target. Inflamm Res 2023; 72:395-423. [PMID: 36565333 PMCID: PMC9789377 DOI: 10.1007/s00011-022-01673-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 07/25/2022] [Accepted: 11/30/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Sepsis has a complex pathogenesis in which the uncontrolled systemic inflammatory response triggered by infection leads to vascular barrier disruption, microcirculation dysfunction and multiple organ dysfunction syndrome. Numerous recent studies reveal that a disintegrin and metalloproteinase 10 (ADAM10) acts as a "molecular scissor" playing a pivotal role in the inflammatory response during sepsis by regulating proteolysis by cleaving various membrane protein substrates, including proinflammatory cytokines, cadherins and Notch, which are involved in intercellular communication. ADAM10 can also act as the cellular receptor for Staphylococcus aureus α-toxin, leading to lethal sepsis. However, its substrate-specific modulation and precise targets in sepsis have not yet to be elucidated. METHODS We performed a computer-based online search using PubMed and Google Scholar for published articles concerning ADAM10 and sepsis. CONCLUSIONS In this review, we focus on the functions of ADAM10 in sepsis-related complex endothelium-immune cell interactions and microcirculation dysfunction through the diversity of its substrates and its enzymatic activity. In addition, we highlight the posttranslational mechanisms of ADAM10 at specific subcellular sites, or in multimolecular complexes, which will provide the insight to intervene in the pathophysiological process of sepsis caused by ADAM10 dysregulation.
Collapse
Affiliation(s)
- Shuanglin Liao
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - Yao Lin
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Lizhen Liu
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - Shuai Yang
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
| | - YingYing Lin
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Junbing He
- The Key Laboratory of Organ Dysfunction and Protection Translational Medicine, Jieyang Medical Research Center, Jieyang People’s Hospital, Tianfu Road 107, Rongcheng District, Jieyang, 522000 Guangdong China
| | - Yiming Shao
- grid.410560.60000 0004 1760 3078The Intensive Care Unit, The First Dongguan Affiliated Hospital, Guangdong Medical University, Jiaoping Road 42, Tangxia Town, Dongguan, 523710 Guangdong China
- grid.410560.60000 0004 1760 3078The Key Laboratory of Sepsis Translational Medicine, Guangdong Medical University, Zhanjiang, Guangdong China
| |
Collapse
|
8
|
de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol Rev 2022; 74:712-768. [PMID: 35738680 DOI: 10.1124/pharmrev.121.000349] [Citation(s) in RCA: 182] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The first matrix metalloproteinase (MMP) was discovered in 1962 from the tail of a tadpole by its ability to degrade collagen. As their name suggests, matrix metalloproteinases are proteases capable of remodeling the extracellular matrix. More recently, MMPs have been demonstrated to play numerous additional biologic roles in cell signaling, immune regulation, and transcriptional control, all of which are unrelated to the degradation of the extracellular matrix. In this review, we will present milestones and major discoveries of MMP research, including various clinical trials for the use of MMP inhibitors. We will discuss the reasons behind the failures of most MMP inhibitors for the treatment of cancer and inflammatory diseases. There are still misconceptions about the pathophysiological roles of MMPs and the best strategies to inhibit their detrimental functions. This review aims to discuss MMPs in preclinical models and human pathologies. We will discuss new biochemical tools to track their proteolytic activity in vivo and ex vivo, in addition to future pharmacological alternatives to inhibit their detrimental functions in diseases. SIGNIFICANCE STATEMENT: Matrix metalloproteinases (MMPs) have been implicated in most inflammatory, autoimmune, cancers, and pathogen-mediated diseases. Initially overlooked, MMP contributions can be both beneficial and detrimental in disease progression and resolution. Thousands of MMP substrates have been suggested, and a few hundred have been validated. After more than 60 years of MMP research, there remain intriguing enigmas to solve regarding their biological functions in diseases.
Collapse
Affiliation(s)
- Luiz G N de Almeida
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Hayley Thode
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Yekta Eslambolchi
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sameeksha Chopra
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Daniel Young
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sean Gill
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Laurent Devel
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Antoine Dufour
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| |
Collapse
|
9
|
Tissue Inhibitor of Metalloproteases 3 (TIMP-3): In Vivo Analysis Underpins Its Role as a Master Regulator of Ectodomain Shedding. MEMBRANES 2022; 12:membranes12020211. [PMID: 35207132 PMCID: PMC8878240 DOI: 10.3390/membranes12020211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 01/06/2023]
Abstract
The proteolytical cleavage of transmembrane proteins with subsequent release of their extracellular domain, so-called ectodomain shedding, is a post-translational modification that plays an essential role in several biological processes, such as cell communication, adhesion and migration. Metalloproteases are major proteases in ectodomain shedding, especially the disintegrin metalloproteases (ADAMs) and the membrane-type matrix metalloproteases (MT-MMPs), which are considered to be canonical sheddases for their membrane-anchored topology and for the large number of proteins that they can release. The unique ability of TIMP-3 to inhibit different families of metalloproteases, including the canonical sheddases (ADAMs and MT-MMPs), renders it a master regulator of ectodomain shedding. This review provides an overview of the different functions of TIMP-3 in health and disease, with a major focus on the functional consequences in vivo related to its ability to control ectodomain shedding. Furthermore, herein we describe a collection of mass spectrometry-based approaches that have been used in recent years to identify new functions of sheddases and TIMP-3. These methods may be used in the future to elucidate the pathological mechanisms triggered by the Sorsby’s fundus dystrophy variants of TIMP-3 or to identify proteins released by less well characterized TIMP-3 target sheddases whose substrate repertoire is still limited, thus providing novel insights into the physiological and pathological functions of the inhibitor.
Collapse
|
10
|
Abu El-Asrar AM, Ahmad A, Nawaz MI, Siddiquei MM, De Zutter A, Vanbrabant L, Gikandi PW, Opdenakker G, Struyf S. Tissue Inhibitor of Metalloproteinase-3 Ameliorates Diabetes-Induced Retinal Inflammation. Front Physiol 2022; 12:807747. [PMID: 35082694 PMCID: PMC8784736 DOI: 10.3389/fphys.2021.807747] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/10/2021] [Indexed: 12/18/2022] Open
Abstract
Purpose: Endogenous tissue inhibitor of matrix metalloproteinase-3 (TIMP-3) has powerful regulatory effects on inflammation and angiogenesis. In this study, we investigated the role of TIMP-3 in regulating inflammation in the diabetic retina. Methods: Vitreous samples from patients with proliferative diabetic retinopathy (PDR) and non-diabetic patients were subjected to Western blot analysis. Streptozotocin-treated rats were used as a preclinical diabetic retinopathy (DR) model. Blood-retinal barrier (BRB) breakdown was assessed with fluorescein isothiocyanate (FITC)-conjugated dextran. Rat retinas, human retinal microvascular endothelial cells (HRMECs) and human retinal Müller glial cells were studied by Western blot analysis and ELISA. Adherence of human monocytes to HRMECs was assessed and in vitro angiogenesis assays were performed. Results: Tissue inhibitor of matrix metalloproteinase-3 in vitreous samples was largely glycosylated. Intravitreal injection of TIMP-3 attenuated diabetes-induced BRB breakdown. This effect was associated with downregulation of diabetes-induced upregulation of the p65 subunit of NF-κB, intercellular adhesion molecule-1 (ICAM-1), and vascular endothelial growth factor (VEGF), whereas phospho-ERK1/2 levels were not altered. In Müller cell cultures, TIMP-3 significantly attenuated VEGF upregulation induced by high-glucose (HG), the hypoxia mimetic agent cobalt chloride (CoCl2) and TNF-α and attenuated MCP-1 upregulation induced by CoCl2 and TNF-α, but not by HG. TIMP-3 attenuated HG-induced upregulation of phospho-ERK1/2, caspase-3 and the mature form of ADAM17, but not the levels of the p65 subunit of NF-κB and the proform of ADAM17 in Müller cells. TIMP-3 significantly downregulated TNF-α-induced upregulation of ICAM-1 and VCAM-1 in HRMECs. Accordingly, TIMP-3 significantly decreased spontaneous and TNF-α- and VEGF-induced adherence of monocytes to HRMECs. Finally, TIMP-3 significantly attenuated VEGF-induced migration, chemotaxis and proliferation of HRMECs. Conclusion:In vitro and in vivo data point to anti-inflammatory and anti-angiogenic effects of TIMP-3 and support further studies for its applications in the treatment of DR.
Collapse
Affiliation(s)
- Ahmed M Abu El-Asrar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Dr. Nasser Al-Rashid Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ajmal Ahmad
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Imtiaz Nawaz
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Alexandra De Zutter
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Lotte Vanbrabant
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Priscilla W Gikandi
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ghislain Opdenakker
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, KU Leuven, and University Hospitals UZ Gasthuisberg, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Jiang J, Ouyang H, Zhou Q, Tang S, Fang P, Xie G, Yang J, Sun G. LPS induces pulmonary microvascular endothelial cell barrier dysfunction by upregulating ceramide production. Cell Signal 2022; 92:110250. [DOI: 10.1016/j.cellsig.2022.110250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/22/2022]
|
12
|
Xu L, Willumeit-Römer R, Luthringer-Feyerabend BJC. Mesenchymal Stem Cell and Oxygen Modulate the Cocultured Endothelial Cells in the Presence of Magnesium Degradation Products. ACS APPLIED BIO MATERIALS 2021; 4:2398-2407. [DOI: 10.1021/acsabm.0c01289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Lei Xu
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | - Regine Willumeit-Römer
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | | |
Collapse
|
13
|
Hypoxia influences the effects of magnesium degradation products on the interactions between endothelial and mesenchymal stem cells. Acta Biomater 2020; 101:624-636. [PMID: 31622779 DOI: 10.1016/j.actbio.2019.10.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023]
Abstract
Biodegradable materials like well-documented Magnesium (Mg) are promising for their biocompatibility and tissue regeneration. Since Mg degradation is reported to be oxygen related, the effects of Mg were hypothesised to be influenced by oxygen. As two vital components of bone marrow, endothelial cells (EC) and mesenchymal stem cells (MSC), their interactions represent high scientific interest for tissue engineering and biodegradable Mg application. Human umbilical cord perivascular (HUCPV) and umbilical vein endothelial cell (HUVEC) were selected as sources of MSC and EC, respectively. Two types of coculture models were established to represent different phases of MSC-EC interaction: (i) where cells were physically separated thanks to a transwell and (ii) where cells were allowed to have heterotypic cellular contacts. Cell migration, gene, cytokines, and proliferation were investigated in HUCPV-HUVEC coculture using DNA, flow cytometry, wound healing assay, semi-quantitative real-time polymerase chain reaction (qRT-PCR), and enzyme-linked immunosorbent assay (ELISA). Mg degradation products increased HUCPV migration in transwell under hypoxia. Oxygen tension changed the gene regulation of migratory, angiogenetic or osteogenic regulators. Under contacting coculture and hypoxia, Mg degradation products remarkably increased cytokines (e.g., c-c motif chemokine ligand 2 and vascular endothelial growth factor) and MSC mineralisation. Mg degradation products decreased and increased the MSC proliferation in transwell and in heterotypic-contact coculture, respectively. In summary, this study indicates the roles of low oxygen and heterotypic contact to effects of Mg materials facilitating HUVEC and HUCPV. STATEMENT OF SIGNIFICANCE.
Collapse
|
14
|
Wang L, Chung J, Gill SE, Mehta S. Quantification of adherens junction disruption and contiguous paracellular protein leak in human lung endothelial cells under septic conditions. Microcirculation 2019; 26:e12528. [PMID: 30636088 DOI: 10.1111/micc.12528] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/12/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Sepsis is associated with dysfunction of MVEC resulting in organ edema and inflammation. VE-cadherin, a component of MVEC adherens junctions, may be disrupted in sepsis. However, the direct connection between individual MVEC VE-cadherin disruption and increased paracellular permeability is uncertain. METHODS Human pulmonary MVEC were cultured on a biotin matrix and treated with cytomix, as a model of sepsis, vs PBS. MVEC permeability was assessed by trans-MVEC monolayer leak of Oregon green 488-conjugated avidin, which bound subcellular biotin to localize sites of paracellular leak. Leak was correlated with individual cell-specific MVEC surface VE-cadherin continuity by fluorescence microscopy. RESULTS Cytomix treatment reduced total MVEC VE-cadherin density, disrupted surface VE-cadherin continuity, was associated with intercellular gap formation, and enhanced paracellular avidin leak. Cytomix-induced MVEC paracellular avidin leak was strongly correlated temporally and was highly contiguous with focal MVEC surface VE-cadherin disruption. Total cellular VE-cadherin density was less strongly correlated with MVEC paracellular avidin leak and individual cell-specific focal surface VE-cadherin discontinuity. CONCLUSIONS These data support a mechanistic link between septic human lung MVEC VE-cadherin disruption and contiguous paracellular protein leak, and will permit more detailed assessment of individual cell-specific mechanisms of septic MVEC barrier dysfunction.
Collapse
Affiliation(s)
- Lefeng Wang
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Division of Respirology, Western University, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada
| | - Justin Chung
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Division of Respirology, Western University, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada.,Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Sanjay Mehta
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Division of Respirology, Western University, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada
| |
Collapse
|
15
|
Hendrickson CM, Gibb SL, Miyazawa BY, Keating SM, Ross E, Conroy AS, Calfee CS, Pati S, Cohen MJ. Elevated plasma levels of TIMP-3 are associated with a higher risk of acute respiratory distress syndrome and death following severe isolated traumatic brain injury. Trauma Surg Acute Care Open 2018; 3:e000171. [PMID: 30023434 PMCID: PMC6045722 DOI: 10.1136/tsaco-2018-000171] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/10/2018] [Indexed: 01/15/2023] Open
Abstract
Background: Complications after injury, such as acute respiratory distress syndrome (ARDS), are common after traumatic brain injury (TBI) and associated with poor clinical outcomes. The mechanisms driving non-neurologic organ dysfunction after TBI are not well understood. Tissue inhibitor of matrix metalloproteinase-3 (TIMP-3) is a regulator of matrix metalloproteinase activity, inflammation, and vascular permeability, and hence has plausibility as a biomarker for the systemic response to TBI. Methods: In a retrospective study of 182 patients with severe isolated TBI, we measured TIMP-3 in plasma obtained on emergency department arrival. We used non-parametric tests and logistic regression analyses to test the association of TIMP-3 with the incidence of ARDS within 8 days of admission and in-hospital mortality. Results: TIMP-3 was significantly higher among subjects who developed ARDS compared with those who did not (median 2810 pg/mL vs. 2260 pg/mL, p=0.008), and significantly higher among subjects who died than among those who survived to discharge (median 2960 pg/mL vs. 2080 pg/mL, p<0.001). In an unadjusted logistic regression model, for each SD increase in plasma TIMP-3, the odds of ARDS increased significantly, OR 1.5 (95% CI 1.1 to 2.1). This association was only attenuated in multivariate models, OR 1.4 (95% CI 1.0 to 2.0). In an unadjusted logistic regression model, for each SD increase in plasma TIMP-3, the odds of death increased significantly, OR 1.7 (95% CI 1.2 to 2.3). The magnitude of this association was greater in a multivariate model adjusted for markers of injury severity, OR 1.9 (95% CI 1.2 to 2.8). Discussion: TIMP-3 may play an important role in the biology of the systemic response to brain injury in humans. Along with clinical and demographic data, early measurements of plasma biomarkers such as TIMP-3 may help identify patients at higher risk of ARDS and death after severe isolated TBI. Level of evidence III.
Collapse
Affiliation(s)
- Carolyn M Hendrickson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Stuart L Gibb
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA.,Blood Systems Research Institute, San Francisco, California, USA
| | - Byron Y Miyazawa
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA.,Blood Systems Research Institute, San Francisco, California, USA.,Department of Surgery, University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Sheila M Keating
- Blood Systems Research Institute, San Francisco, California, USA
| | - Erin Ross
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Amanda S Conroy
- Department of Surgery, University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Carolyn S Calfee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Shibani Pati
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA.,Blood Systems Research Institute, San Francisco, California, USA
| | - Mitchell J Cohen
- Department of Surgery, University of California San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, California, USA.,Department of Surgery, University of Colorado, Denver, Colorado, USA
| |
Collapse
|
16
|
Simvastatin Ameliorates PAK4 Inhibitor-Induced Gut and Lung Injury. BIOMED RESEARCH INTERNATIONAL 2018; 2017:8314276. [PMID: 29445744 PMCID: PMC5763212 DOI: 10.1155/2017/8314276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 11/29/2017] [Indexed: 11/17/2022]
Abstract
P21 activated kinase 4 (PAK4), a key regulator of cytoskeletal rearrangement and endothelial microparticles (EMPs), is released after lipopolysaccharide (LPS) stimulation. In addition, it participates in LPS-induced lung injury. In this study, forty-eight Sprague Dawley (SD) rats were divided into two groups, including PAK4 inhibitor (P) and PAK4 inhibitor + simvastatin (P + S) treatment groups. All rats were given PAK4 inhibitor (15 mg/kg/d) orally. Immediately after PAK4 inhibitor administration, simvastatin was injected intraperitoneally to P + S group animals at 20 mg/kg/day. Then, treatment effects on the intestinal mucosal barrier and lung injury caused by PAK4 inhibitor and simvastatin were assessed. The results showed that gut Zonula Occludens- (ZO-) 1, PAK4, mitogen-activated protein kinase 4 (MPAK4), and CD11c protein levels were reduced, while plasma endotoxin levels were increased after administration of PAK4 inhibitor. Furthermore, compared with normal rats, wet-to-dry (W/D) values of lung tissues and circulating EMP levels were increased in the treatment group, while PAK4 and CD11c protein amounts were reduced. Therefore, in this lung injury process induced by PAK4 inhibitor, the protective effects of simvastatin were reflected by intestinal mucosal barrier protection, inflammatory response regulation via CD11c+ cells, and cytoskeleton stabilization. In summary, PAK4 is a key regulator in the pathophysiological process of acute lung injury (ALI) and can be a useful target for ALI treatment.
Collapse
|
17
|
Simvastatin Attenuates Acute Lung Injury via Regulating CDC42-PAK4 and Endothelial Microparticles. Shock 2018; 47:378-384. [PMID: 27513084 DOI: 10.1097/shk.0000000000000723] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Simvastatin has lung vascular-protective effects via augmentation of endothelial barrier function. Accordingly, on the basis of our previous study, we hypothesized that endothelial cell (EC) protection by simvastatin is dependent on the stabilization on cytoskeletons. METHODS Sixty C57BL/6 mice were divided into two experimental groups: lipopolysaccharide (LPS) group (L group) and LPS+simvastatin treated group (L+S group). All mice in these two groups received an intraperitoneal injection of LPS (10 mg/kg/d). Simvastatin was administered intraperitoneally immediately after the LPS injection in animals of the L+S group at a dose of 20 mg/kg/day. Lung injury degree and the protective effects of simvastatin against LPS-induced lung injury were assessed at the time-points of 24, 48, and 72 h postinjection. Serum alanine transaminase (ALT), serum creatinine (Scr) were identified to assess the hepatic and renal side-effects of simvastatin. RESULTS LPS inhibited the cytoskeletal regulating proteins of Cdc42 and PAK4, and was accompanied by an increased circulating endothelial microparticles (EMPs) level. The adherent junction (AJ) protein of VE-cadherin was also decreased by LPS, and was accompanied by a thickening alveolar wall, increased lung W/D values, and high albumin concentration in bronchoalveolar lavage. Protective effects of simvastatin against LPS-induced lung injury were illustrated by regulating and stabilizing cytoskeletons, as well as intercellular AJs. The values of ALT and Scr were all lower than the common upper limits according to assay kits. CONCLUSION An increased serous EMP level associated with Cdc42-PAK4 can be deemed as a useful pulmonary injury marker in LPS-treated mice, and our results might be more relevant in guiding the clinical treatment of ALI by intervening Cdc42-PAK4 or EMPs.
Collapse
|
18
|
Dai J, Chen W, Lin Y, Wang S, Guo X, Zhang QQ. Exposure to Concentrated Ambient Fine Particulate Matter Induces Vascular Endothelial Dysfunction via miR-21. Int J Biol Sci 2017; 13:868-877. [PMID: 28808419 PMCID: PMC5555104 DOI: 10.7150/ijbs.19868] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/03/2017] [Indexed: 12/22/2022] Open
Abstract
Vascular endothelial permeability transition does not cause significant lesions, but enhanced permeability may contribute to the development of vascular and other diseases, including atherosclerosis, hypertension, heart failure and cancer. Therefore, elucidating the effect of Particulate Matter 2.5 (PM2.5) on vascular endothelial permeability could help prevent disease that might be caused by PM2.5. Our previous study and the present one revealed that PM2.5 significantly increased the permeability of vascular endothelial cells and disrupted the barrier function of the vascular endothelium in Sprague Dawley (SD) rats. We found that the effect occurred mainly through induction of signal transducer and activator of transcription 3 (STAT3) phosphorylation, further transcriptional regulation of microRNA21 (miR-21) and promotion of miR-21 expression. These changes post-transcriptionally repress tissue inhibitor of metalloproteinases 3 (TIMP3) and promote matrix metalloproteinases 9 (MMP9) expression. This work provides evidence that PM2.5 exerts direct inhibitory action on vascular endothelial barrier function and might give rise to a number of vascular diseases.
Collapse
Affiliation(s)
- Jianwei Dai
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 510182, China.,The State Key Lab of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China
| | - Wensheng Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 510182, China
| | - Yuyin Lin
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 510182, China
| | - Shiwen Wang
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 510182, China
| | - Xiaolan Guo
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 510182, China
| | - Qian-Qian Zhang
- Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
19
|
Roubinian NH, Looney MR, Keating S, Kor DJ, Lowell CA, Gajic O, Hubmayr R, Gropper M, Koenigsberg M, Wilson GA, A Matthay M, Toy P, Murphy EL. Differentiating pulmonary transfusion reactions using recipient and transfusion factors. Transfusion 2017; 57:1684-1690. [PMID: 28470756 DOI: 10.1111/trf.14118] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/07/2017] [Accepted: 02/12/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND It is increasingly recognized that recipient risk factors play a prominent role in possible transfusion-related acute lung injury (pTRALI) and transfusion-associated circulatory overload (TACO). We hypothesized that both transfusion and recipient factors including natriuretic peptides could be used to distinguish TRALI from TACO and pTRALI. STUDY DESIGN AND METHODS We performed a post hoc analysis of a case-control study of pulmonary transfusion reactions conducted at the University of California at San Francisco and Mayo Clinic, Rochester. We evaluated clinical data and brain natriuretic peptides (BNP) levels drawn after transfusion in patients with TRALI (n = 21), pTRALI (n = 26), TACO (n = 22), and controls (n = 24). Logistic regression and receiver operating characteristics curve analyses were used to determine the accuracy of clinical and biomarker predictors in differentiating TRALI from TACO and pTRALI. RESULTS We found that pTRALI and TACO were associated with older age, higher fluid balance, and elevated BNP levels relative to those of controls and TRALI. The following variables were useful in distinguishing cases of pTRALI and TACO from TRALI: age more than 70 years, BNP levels more than 1000 pg/mL, 24-hour fluid balance of more than 3 L, and a lower number of transfused blood components. Using the above variables, our logistic model had a 91% negative predictive value in the differential diagnosis of TRALI. CONCLUSIONS Models incorporating readily available clinical and biomarker data can be used to differentiate transfusion-related respiratory complications. Additional studies examining recipient risk factors and the likelihood of TRALI may be useful in decision making regarding donor white blood cell antibody testing.
Collapse
Affiliation(s)
- Nareg H Roubinian
- Blood Systems Research Institute.,University of California at San Francisco, San Francisco, California
| | - Mark R Looney
- University of California at San Francisco, San Francisco, California
| | | | | | | | | | | | - Michael Gropper
- University of California at San Francisco, San Francisco, California
| | | | | | - Michael A Matthay
- University of California at San Francisco, San Francisco, California
| | - Pearl Toy
- University of California at San Francisco, San Francisco, California
| | - Edward L Murphy
- Blood Systems Research Institute.,University of California at San Francisco, San Francisco, California
| | | |
Collapse
|
20
|
Masciantonio MG, Lee CKS, Arpino V, Mehta S, Gill SE. The Balance Between Metalloproteinases and TIMPs: Critical Regulator of Microvascular Endothelial Cell Function in Health and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:101-131. [PMID: 28413026 DOI: 10.1016/bs.pmbts.2017.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cells (EC), especially the microvascular EC (MVEC), have critical functions in health and disease. For example, healthy MVEC provide a barrier between the fluid and protein found within the blood, and the surrounding tissue. Following tissue injury or infection, the microvascular barrier is often disrupted due to activation and dysfunction of the MVEC. Multiple mechanisms promote MVEC activation and dysfunction, including stimulation by cytokines, mechanical interaction with activated leukocytes, and exposure to harmful leukocyte-derived molecules, which collectively result in a loss of MVEC barrier function. However, MVEC activation is also critical to facilitate recruitment of inflammatory cells, such as neutrophils (PMNs) and monocytes, into the injured or infected tissue. Metalloproteinases, including the matrix metalloproteinases (MMPs) and the closely related, a disintegrin and metalloproteinases (ADAMs), have been implicated in regulating both MVEC barrier function, through cleavage of adherens and tight junctions proteins between adjacent MVEC and through degradation of the extracellular matrix, as well as PMN-MVEC interaction, through shedding of cell surface PMN receptors. Moreover, the tissue inhibitors of metalloproteinases (TIMPs), which collectively inhibit most MMPs and ADAMs, are critical regulators of MVEC activation and dysfunction through their ability to inhibit metalloproteinases and thereby promote MVEC stability. However, TIMPs have been also found to modulate MVEC function through metalloproteinase-independent mechanisms, such as regulation of vascular endothelial growth factor signaling. This chapter is focused on examining the role of the metalloproteinases and TIMPs in regulation of MVEC function in both health and disease.
Collapse
Affiliation(s)
- Marcello G Masciantonio
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Christopher K S Lee
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Valerie Arpino
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Sanjay Mehta
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada; Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| |
Collapse
|
21
|
Inhibition of Murine Pulmonary Microvascular Endothelial Cell Apoptosis Promotes Recovery of Barrier Function under Septic Conditions. Mediators Inflamm 2017; 2017:3415380. [PMID: 28250575 PMCID: PMC5303866 DOI: 10.1155/2017/3415380] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/25/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022] Open
Abstract
Sepsis is characterized by injury of the pulmonary microvasculature and the pulmonary microvascular endothelial cells (PMVEC), leading to barrier dysfunction and acute respiratory distress syndrome (ARDS). Our recent work identified a strong correlation between PMVEC apoptosis and microvascular leak in septic mice in vivo, but the specific role of apoptosis in septic PMVEC barrier dysfunction remains unclear. Thus, we hypothesize that PMVEC apoptosis is likely required for PMVEC barrier dysfunction under septic conditions in vitro. Septic stimulation (mixture of tumour necrosis factor α, interleukin 1β, and interferon γ [cytomix]) of isolated murine PMVEC resulted in a significant loss of barrier function as early as 4 h after stimulation, which persisted until 24 h. PMVEC apoptosis, as reflected by caspase activation, DNA fragmentation, and loss of membrane polarity, was first apparent at 8 h after cytomix. Pretreatment of PMVEC with the pan-caspase inhibitor Q-VD significantly decreased septic PMVEC apoptosis and was associated with reestablishment of PMVEC barrier function at 16 and 24 h after stimulation but had no effect on septic PMVEC barrier dysfunction over the first 8 h. Collectively, our data suggest that early septic murine PMVEC barrier dysfunction driven by proinflammatory cytokines is not mediated through apoptosis, but PMVEC apoptosis contributes to late septic PMVEC barrier dysfunction.
Collapse
|
22
|
Zielińska KA, Van Moortel L, Opdenakker G, De Bosscher K, Van den Steen PE. Endothelial Response to Glucocorticoids in Inflammatory Diseases. Front Immunol 2016; 7:592. [PMID: 28018358 PMCID: PMC5155119 DOI: 10.3389/fimmu.2016.00592] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/29/2016] [Indexed: 12/16/2022] Open
Abstract
The endothelium plays a crucial role in inflammation. A balanced control of inflammation requires the action of glucocorticoids (GCs), steroidal hormones with potent cell-specific anti-inflammatory properties. Besides the classic anti-inflammatory effects of GCs on leukocytes, recent studies confirm that endothelial cells also represent an important target for GCs. GCs regulate different aspects of endothelial physiology including expression of adhesion molecules, production of pro-inflammatory cytokines and chemokines, and maintenance of endothelial barrier integrity. However, the regulation of endothelial GC sensitivity remains incompletely understood. In this review, we specifically examine the endothelial response to GCs in various inflammatory diseases ranging from multiple sclerosis, stroke, sepsis, and vasculitis to atherosclerosis. Shedding more light on the cross talk between GCs and endothelium will help to improve existing therapeutic strategies and develop new therapies better tailored to the needs of patients.
Collapse
Affiliation(s)
- Karolina A. Zielińska
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Laura Van Moortel
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-UGent, VIB Medical Biotechnology Center, Ghent, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-UGent, VIB Medical Biotechnology Center, Ghent, Belgium
| | | |
Collapse
|
23
|
Lung remodeling associated with recovery from acute lung injury. Cell Tissue Res 2016; 367:495-509. [DOI: 10.1007/s00441-016-2521-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022]
|
24
|
Mandel ER, Uchida C, Nwadozi E, Makki A, Haas TL. Tissue Inhibitor of Metalloproteinase 1 Influences Vascular Adaptations to Chronic Alterations in Blood Flow. J Cell Physiol 2016; 232:831-841. [PMID: 27430487 DOI: 10.1002/jcp.25491] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 07/18/2016] [Indexed: 12/22/2022]
Abstract
Remodeling of the skeletal muscle microvasculature involves the coordinated actions of matrix metalloproteinases (MMPs) and their endogenous inhibitors, tissue inhibitor of metalloproteinases (TIMPs). We hypothesized that the loss of TIMP1 would enhance both ischemia and flow-induced vascular remodeling by increasing MMP activity. TIMP1 deficient (Timp1-/- ) and wild-type (WT) C57BL/6 mice underwent unilateral femoral artery (FA) ligation or were treated with prazosin, an alpha-1 adrenergic receptor antagonist, in order to investigate vascular remodeling to altered flow. Under basal conditions, Timp1-/- mice had reduced microvascular content as compared to WT mice. Furthermore, vascular remodeling was impaired in Timp1-/- mice. Timp1-/- mice displayed reduced blood flow recovery in response to FA ligation and no arteriogenic response to prazosin treatment. Timp1-/- mice failed to undergo angiogenesis in response to ischemia or prazosin, despite maintaining the capacity to increase VEGF-A and eNOS mRNA. Vascular permeability was increased in muscles of Timp1-/- mice in response to both prazosin treatment and FA ligation, but this was not accompanied by greater MMP activity. This study highlights a previously undescribed integral role for TIMP1 in both vascular network maturation and adaptations to ischemia or alterations in flow. J. Cell. Physiol. 232: 831-841, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Erin R Mandel
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Cassandra Uchida
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Emmanuel Nwadozi
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Armin Makki
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Tara L Haas
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| |
Collapse
|