1
|
Liu C, Zhang J, Qi B, Li Z, Li Q, Wang L, Li C, Wang Z, Qi Z, Lu C. Deficiency of Caveolin-1 Aggravates Cardiac Infarction Injury by Disturbing the Endothelial Homeostasis. Int J Med Sci 2025; 22:920-932. [PMID: 39991756 PMCID: PMC11843147 DOI: 10.7150/ijms.101074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/19/2024] [Indexed: 02/25/2025] Open
Abstract
Objective: Caveolin-1 (Cav-1) plays a crucial role in maintaining the homeostasis of vascular endothelium. Endothelial dysfunction is involved in many ischemic diseases. However, the role of Cav-1 in myocardial infarction (MI) has not been fully elucidated. Here, this study aims to delineate the function of Cav-1 in MI injury and its impact on endothelial homeostasis. Methods: To elucidate the role of Cav-1 in MI in vivo, we generated the global knockout Cav-1 (Cav-1-KO) mice. And we manipulated the expression of Cav-1 by siRNA in vitro to evaluate the effects apoptosis, inflammatory response and oxidative stress as well as autophagy flux under the hypoxic model of endothelial cells (ECs). Results: Initially, we found that Cav-1 was mainly expressed in vascular endothelial cells of myocardium. Interestingly, we found that Cav-1 deficiency significantly amplified the size of myocardial infarction areas, alongside the deteriorated cardiac function in vivo. Consistently in vitro, siRNA-mediated knockdown of Cav-1 exacerbated the endothelial apoptosis, inflammatory response and oxidative stress as well as eliminated autophagy flux. However, pretreatment with β-cyclodextrin (β-CD), which depletes membrane-bound cholesterol and disrupts lipid rafts, markedly mitigated the effects induced by downregulation of Cav-1, respectively. Conclusion: Collectively, in this study, we demonstrated that Cav-1 acts as a protective regulator of MI injury through maintaining endothelial homeostasis. These findings implied that Cav-1 may be a potential therapeutic target for MI injury.
Collapse
Affiliation(s)
- Chunlei Liu
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
- Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Junwei Zhang
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Bing Qi
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhuqing Li
- Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Qi Li
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443003, China
| | - Li Wang
- Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Chao Li
- Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Ziwei Wang
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Zhi Qi
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Chengzhi Lu
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
| |
Collapse
|
2
|
Johnson CF, Schafer CM, Burge KY, Coon BG, Chaaban H, Griffin CT. Endothelial RIPK3 minimizes organotypic inflammation and vascular permeability in ischemia-reperfusion injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625188. [PMID: 39651150 PMCID: PMC11623548 DOI: 10.1101/2024.11.25.625188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Recent studies have revealed a link between endothelial receptor-interacting protein kinase 3 (RIPK3) and vascular integrity. During mouse embryonic development, hypoxia can trigger elevated endothelial RIPK3 that contributes to lethal vascular rupture. However, it is unknown whether RIPK3 regulate endothelial barrier function in adult vasculature under hypoxic injury conditions such as ischemia-reperfusion (I/R) injury. Here we performed inducible genetic deletion of endothelial Ripk3 ( Ripk iECKO ) in mice, which led to elevated vascular permeability in the small intestine and multiple distal organs after intestinal I/R injury. Mechanistically, this vascular permeability correlated with increased endothelial secretion of IL-6 and organ-specific expression of VCAM-1 and ICAM-1 adhesion molecules. Circulating monocyte depletion with clodronate liposomes reduced permeability in organs with elevated adhesion molecules, highlighting the contribution of monocyte adhesion and extravasation to Ripk iECKO barrier dysfunction. These results elucidate mechanisms by which RIPK3 regulates endothelial inflammation to minimize vascular permeability in I/R injury. GRAPHICAL ABSTRACT
Collapse
|
3
|
Song P, Xu Y, Ye G. B7-H3 and ICAM-1 are potentially therapeutic targets for thyroid carcinoma. Diagn Pathol 2024; 19:77. [PMID: 38858715 PMCID: PMC11163747 DOI: 10.1186/s13000-024-01504-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024] Open
Abstract
Although most differentiated thyroid carcinoma has a clinically favorable prognosis, some of specific types of thyroid cancer (such as anaplastic thyroid carcinoma and advanced papillary thyroid carcinoma) show fatal outcomes and require novel treatments. Immunotherapy is a promising avenue for the treatment of advanced thyroid carcinoma. B7-H3 (B7 homolog 3 protein) and ICAM-1 (intercellular adhesion molecule 1), as two important immune checkpoints (ICPs), is becoming hopeful target spots for immunotherapy. A growing amount of evidence has suggested that B7-H3 and ICAM-1 are upregulated in papillary thyroid carcinoma. However, their expression level in specific types of thyroid cancer remains largely unclear. In the present study, we explored the expression level of B7-H3 and ICAM-1 in different types of thyroid carcinoma. In the groups of the TCGA cohort, both B7-H3 and ICAM-1 mRNA were highly expressed in thyroid carcinoma. Furthermore, the patients with Stage2, 61-80y, Follicular thyroid papillary carcinoma and N0 had lower B7-H3 and ICAM-1 mRNA expression. In the groups of our cohort, PTCs and ATCs showed frequently moderate to strong expression of B7-H3 and ICAM-1 protein expression. The significant relevance of B7-H3 staining score with ICAM-1 staining score was observed in TCGA database and our cohort, which might open avenues for the combination therapy in advanced thyroid cancer.
Collapse
Affiliation(s)
- Pengtao Song
- Department of Pathology, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China
- Department of Pathology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China
| | - Yongcan Xu
- Department of General Surgery, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China.
- Department of General Surgery, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, 313000, People's Republic of China.
| | - Guochao Ye
- Department of General Surgery, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China.
- Department of General Surgery, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, 313000, People's Republic of China.
| |
Collapse
|
4
|
Lin S, Zhu P, Jiang L, Hu Y, Huang L, He Y, Zhang H. Neutrophil extracellular traps induced by IL-1β promote endothelial dysfunction and aggravate limb ischemia. Hypertens Res 2024; 47:1654-1667. [PMID: 38605142 DOI: 10.1038/s41440-024-01661-3] [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: 10/17/2023] [Revised: 02/22/2024] [Accepted: 03/13/2024] [Indexed: 04/13/2024]
Abstract
Vascular inflammation and endothelial dysfunction contribute to vascular diseases. While neutrophil extracellular traps (NETs) participate in some vascular pathologies, their roles in lower limb ischemia remain poorly defined. This study investigated the functional significance of NETs in vascular inflammation and remodeling associated with limb ischemia. Single-cell RNA sequencing (scRNA-seq) and flow cytometry revealed neutrophil activation and upregulated NETs formation in human limb ischemia, with immunofluorescence confirming IL-1β-induced release of NETs for vascular inflammation. Endothelial cell activation was examined via scRNA-seq and western blotting, indicating enhanced proliferation, expression of adhesion molecules (VCAM-1, ICAM-1), inflammatory cytokines (IL-1β, IL-6) and decreased expression of VE-cadherin, that could be mediated by NETs to exacerbate endothelial inflammation. Mechanistically, NETs altered endothelial cell function via increased pSTAT1/STAT1 signaling. Vascular inflammation and subsequent ischemia were alleviated in vivo by NETosis or IL-1β inhibition in ischemic mice. IL-1β-NETs induce endothelial activation and inflammation in limb ischemia by stimulating STAT1 signaling. Targeting NETs may thus represent a novel therapeutic strategy for inflammatory vascular diseases associated with limb ischemia. Graphical abstract of NETs regulation of the development of vascular inflammation in lower limb ischemia via pSTAT1/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Shigang Lin
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengwei Zhu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yujian Hu
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lirui Huang
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yangyan He
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Hongkun Zhang
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Trevino TN, Fogel AB, Minshall R, Richner JM, Lutz SE. Caveolin-1 mediates neuroinflammation and cognitive impairment in SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.563024. [PMID: 37905019 PMCID: PMC10614946 DOI: 10.1101/2023.10.18.563024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Leukocyte infiltration of the CNS can contribute to neuroinflammation and cognitive impairment. Brain endothelial cells regulate adhesion, activation, and diapedesis of T cells across the blood-brain barrier (BBB) in inflammatory diseases. The integral membrane protein Caveolin-1 (Cav-1) critically regulates BBB permeability, but its influence on T cell CNS infiltration in respiratory viral infections is unknown. In this study, we sought to determine the role of Cav-1 at the BBB in neuroinflammation in a COVID-19 mouse model. We used mice genetically deficient in Cav-1 to test the role of this protein in T cell infiltration and cognitive impairment. We found that SARS-CoV-2 infection upregulated brain endothelial Cav-1. Moreover, SARS-CoV-2 infection increased brain endothelial cell vascular cell adhesion molecule-1 (VCAM-1) and CD3+ T cell infiltration of the hippocampus, a region important for short term learning and memory. Concordantly, we observed learning and memory deficits. Importantly, genetic deficiency in Cav-1 attenuated brain endothelial VCAM-1 expression and T cell infiltration in the hippocampus of mice with SARS-CoV-2 infection. Moreover, Cav-1 KO mice were protected from the learning and memory deficits caused by SARS-CoV-2 infection. These results indicate the importance of BBB permeability in COVID-19 neuroinflammation and suggest potential therapeutic value of targeting Cav-1 to improve disease outcomes.
Collapse
|
6
|
Liang C, Zhu D, Xia W, Hong Z, Wang QS, Sun Y, Yang YC, Han SQ, Tang LL, Lou J, Wu MM, Zhang ZR. Inhibition of YAP by lenvatinib in endothelial cells increases blood pressure through ferroptosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166586. [PMID: 36374802 DOI: 10.1016/j.bbadis.2022.166586] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2022]
Abstract
Lenvatinib, a multitarget tyrosine kinase inhibitor (TKI), increases the incidence of severe hypertension and thus the incidence of cardiovascular complications. Inhibition of ferroptosis, a newly recognized type of cell death, alleviates endothelial dysfunction. Here, we report that lenvatinib-induced hypertension is associated with ferroptosis of endothelial cells. RNA sequencing (RNA-seq) showed that lenvatinib led to ferroptosis of endothelial cells and that administration of mouse with ferrostatin-1 (Fer-1), a specific ferroptosis inhibitor, dramatically ameliorated lenvatinib-induced hypertension and reversed lenvatinib-induced impairment of endothelium-dependent relaxation (EDR). Furthermore, lenvatinib significantly reduced glutathione peroxidase 4 (GPX4) expressions in the mouse aorta and human umbilical vein endothelial cells (HUVECs) and increased lipid peroxidation, lactate dehydrogenase (LDH) release, and malondialdehyde (MDA) levels in HUVECs. Immunofluorescence and Western blotting showed that lenvatinib significantly reduced Yes-associated protein (YAP) nuclear translocation but not cytoplasmic YAP expression in HUVECs. The data, generated from both in vivo and in vitro, showed that lenvatinib reduced total YAP (t-YAP) expression and increased the phosphorylation of YAP at both Ser127 and Ser397, without affecting YAP mRNA levels in HUVECs. XMU-MP-1 mediated YAP activation or YAP overexpression effectively attenuated the lenvatinib-induced decrease in GPX4 expression and increases in LDH release and MDA levels. In addition, overexpression of YAP in HUVECs ameliorated lenvatinib-induced decrease in the mRNA and protein levels of spermidine/spermine N (1)-acetyltransferase-1 (SAT1), heme oxygenase-1 (HO-1), and ferritin heavy chain 1 (FTH1). Taken together, our data suggest that lenvatinib-induced inhibition of YAP led to ferroptosis of endothelial cells and subsequently resulted in vascular dysfunction and hypertension.
Collapse
Affiliation(s)
- Chen Liang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Di Zhu
- Departments of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Harbin Medical University, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Wei Xia
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Zi Hong
- Departments of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Harbin Medical University, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Qiu-Shi Wang
- Departments of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Harbin Medical University, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Yu Sun
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Yan-Chao Yang
- Departments of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Harbin Medical University, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Song-Qi Han
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Liang-Liang Tang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Jie Lou
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Ming-Ming Wu
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China
| | - Zhi-Ren Zhang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, PR China; Departments of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Harbin Medical University, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, PR China.
| |
Collapse
|
7
|
Regev O, Kizner M, Roncato F, Dadiani M, Saini M, Castro-Giner F, Yajuk O, Kozlovski S, Levi N, Addadi Y, Golani O, Ben-Dor S, Granot Z, Aceto N, Alon R. ICAM-1 on Breast Cancer Cells Suppresses Lung Metastasis but Is Dispensable for Tumor Growth and Killing by Cytotoxic T Cells. Front Immunol 2022; 13:849701. [PMID: 35911772 PMCID: PMC9328178 DOI: 10.3389/fimmu.2022.849701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Breast tumors and their derived circulating cancer cells express the leukocyte β2 integrin ligand Intercellular adhesion molecule 1 (ICAM-1). We found that elevated ICAM-1 expression in breast cancer cells results in a favorable outcome and prolonged survival of breast cancer patients. We therefore assessed the direct in vivo contribution of ICAM-1 expressed by breast cancer cells to breast tumorigenesis and lung metastasis in syngeneic immunocompetent mice hosts using spontaneous and experimental models of the lung metastasis of the C57BL/6-derived E0771 cell line, a luminal B breast cancer subtype. Notably, the presence of ICAM-1 on E0771 did not alter tumor growth or the leukocyte composition in the tumor microenvironment. Interestingly, the elimination of Tregs led to the rapid killing of primary tumor cells independently of tumor ICAM-1 expression. The in vivo elimination of a primary E0771 tumor expressing the ovalbumin (OVA) model neoantigen by the OVA-specific OVA-tcr-I mice (OT-I) transgenic cytotoxic T lymphocytes (CTLs) also took place normally in the absence of ICAM-1 expression by E0771 breast cancer target cells. The whole lung imaging of these cells by light sheet microscopy (LSM) revealed that both Wild type (WT)- and ICAM-1-deficient E0771 cells were equally disseminated from resected tumors and accumulated inside the lung vasculature at similar magnitudes. ICAM-1-deficient breast cancer cells developed, however, much larger metastatic lesions than their control counterparts. Strikingly, the vast majority of these cells gave rise to intravascular tumor colonies both in spontaneous and experimental metastasis models. In the latter model, ICAM-1 expressing E0771- but not their ICAM-1-deficient counterparts were highly susceptible to elimination by neutrophils adoptively transferred from E0771 tumor-bearing donor mice. Ex vivo, neutrophils derived from tumor-bearing mice also killed cultured E0771 cells via ICAM-1-dependent interactions. Collectively, our results are a first indication that ICAM-1 expressed by metastatic breast cancer cells that expand inside the lung vasculature is involved in innate rather than in adaptive cancer cell killing. This is also a first indication that the breast tumor expression of ICAM-1 is not required for CTL-mediated killing but can function as a suppressor of intravascular breast cancer metastasis to lungs.
Collapse
Affiliation(s)
- Ofer Regev
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Marina Kizner
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Francesco Roncato
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Maya Dadiani
- Cancer Research Center, Sheba Medical Center, Ramat-Gan, Israel
| | - Massimo Saini
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Francesc Castro-Giner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Olga Yajuk
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem, Israel
| | - Stav Kozlovski
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Nehora Levi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Shifra Ben-Dor
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Zvi Granot
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem, Israel
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
8
|
Jones JH, Minshall RD. Endothelial Transcytosis in Acute Lung Injury: Emerging Mechanisms and Therapeutic Approaches. Front Physiol 2022; 13:828093. [PMID: 35431977 PMCID: PMC9008570 DOI: 10.3389/fphys.2022.828093] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/28/2022] [Indexed: 01/08/2023] Open
Abstract
Acute Lung Injury (ALI) is characterized by widespread inflammation which in its severe form, Acute Respiratory Distress Syndrome (ARDS), leads to compromise in respiration causing hypoxemia and death in a substantial number of affected individuals. Loss of endothelial barrier integrity, pneumocyte necrosis, and circulating leukocyte recruitment into the injured lung are recognized mechanisms that contribute to the progression of ALI/ARDS. Additionally, damage to the pulmonary microvasculature by Gram-negative and positive bacteria or viruses (e.g., Escherichia coli, SARS-Cov-2) leads to increased protein and fluid permeability and interstitial edema, further impairing lung function. While most of the vascular leakage is attributed to loss of inter-endothelial junctional integrity, studies in animal models suggest that transendothelial transport of protein through caveolar vesicles, known as transcytosis, occurs in the early phase of ALI/ARDS. Here, we discuss the role of transcytosis in healthy and injured endothelium and highlight recent studies that have contributed to our understanding of the process during ALI/ARDS. We also cover potential approaches that utilize caveolar transport to deliver therapeutics to the lungs which may prevent further injury or improve recovery.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,Department of Anesthesiology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,*Correspondence: Richard D. Minshall,
| |
Collapse
|
9
|
Xu BW, Cheng ZQ, Zhi XT, Yang XM, Yan ZB. Effect of p18 on endothelial barrier function by mediating vascular endothelial Rab11a-VE-cadherin recycling. Biosci Biotechnol Biochem 2021; 85:2392-2403. [PMID: 34747973 DOI: 10.1093/bbb/zbab172] [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: 07/22/2021] [Accepted: 08/26/2021] [Indexed: 11/14/2022]
Abstract
Endothelial barrier integrity requires recycling of VE-cadherin to adherens junctions. Both p18 and Rab11a play significant roles in VE-cadherin recycling. However, the underlying mechanism and the role of p18 in activating Rab11a have yet to be elucidated. Performing in vitro and in vivo experiments, we showed that p18 protein bound to VE-cadherin before Rab11a through its VE-cadherin-binding domain (aa 1-39). Transendothelial resistance showed that overexpression of p18 promoted the circulation of VE-cadherin to adherens junctions and the recovery of the endothelial barrier. Silencing of p18 caused endothelial barrier dysfunction and prevented Rab11a-positive recycling endosome accumulation in the perinuclear recycling compartments. Furthermore, p18 knockdown in pulmonary microvessels markedly increased vascular leakage in mice challenged with lipopolysaccharide and cecal ligation puncture. This study showed that p18 regulated the pulmonary endothelial barrier function in vitro and in vivo by regulating the binding of Rab11a to VE-cadherin and the activation of Rab11a.
Collapse
Affiliation(s)
- Bo-Wen Xu
- Department of Hepatobiliary Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Zhi-Qiang Cheng
- Department of Colorectal Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xu-Ting Zhi
- Department of Hepatobiliary Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiao-Mei Yang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Zhi-Bo Yan
- Department of Colorectal Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
10
|
Chen L, Xu J, Deng M, Liang Y, Ma J, Zhang L, Wang Y, Zhang J. Telmisartan mitigates lipopolysaccharide (LPS)-induced production of mucin 5AC (MUC5AC) through increasing suppressor of cytokine signaling 1 (SOCS1). Bioengineered 2021; 12:3912-3923. [PMID: 34281463 PMCID: PMC8806622 DOI: 10.1080/21655979.2021.1943605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute lung injury (ALI) is a serious clinical pulmonary disease. The pathogenesis of ALI is related to the excessive release of inflammatory factors and upregulation of mucin 5AC (MUC5AC). Telmisartan is a novel antihypertension agent that exerts promising anti-inflammatory effects. The purpose of this study is to investigate whether Telmisartan has a protective role in lipopolysaccharide (LPS)-induced MUC5AC expression and to explore the underlying mechanism in human bronchial epithelial cells. Firstly, the decreased cell viability, elevated release of lactate dehydrogenase (LDH), and excessively released inflammatory factors tumor necrosis factor-α (TNF-α), interleukin- 6 (IL-6), and transforming growth factor-β (TGF)-β in bronchial BEAS-2B epithelial cells induced by stimulation with LPS were significantly reversed by the introduction of Telmisartan. Secondly, the upregulated MUC5AC and downregulated suppressor of cytokine signaling 1 (SOCS1) caused by stimulation with LPS were dramatically reversed by Telmisartan. Notably, treatment with Telmisartan attenuated LPS-induced activation of nuclear factor κ-B (NF-κB). Lastly, silencing of SOCS1 abolished the protective effects of Telmisartan against LPS-induced production of MUC5AC and the activation of NF-κB. Based on these findings, we conclude that Telmisartan displayed a protective effect against LPS by improving mitochondrial function, mitigating inflammatory response, and reducing the production of mucin 5AC by regulating the SOCS1/NF-κB axis in human bronchial epithelial cells.
Collapse
Affiliation(s)
- Ling Chen
- Department of Respiration, Hospital of PLA, Beijing, China
| | - Jiajia Xu
- Department of Pathology, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
| | - Meiyu Deng
- Department of Respiration, Hospital of PLA, Beijing, China
| | - Yanling Liang
- Department of Endocrinology, Hospital of PLA, Beijing, China
| | - Jinfu Ma
- Department of Respiration, Hospital of PLA, Beijing, China
| | - Linghui Zhang
- Department of Respiration, Hospital of PLA, Beijing, China
| | - Yijie Wang
- Department of Respiration, Hospital of PLA, Beijing, China
| | - Jinping Zhang
- Department of Gerontology, Hospital of PLA, Beijing, China
| |
Collapse
|
11
|
Vishnevskiy DA, Garanina AS, Chernysheva AA, Chekhonin VP, Naumenko VA. Neutrophil and Nanoparticles Delivery to Tumor: Is It Going to Carry That Weight? Adv Healthc Mater 2021; 10:e2002071. [PMID: 33734620 DOI: 10.1002/adhm.202002071] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/02/2021] [Indexed: 12/15/2022]
Abstract
The application of cell carriers for transporting nanodrugs to the tumor draws much attention as the alternative to the passive drug delivery. In this concept, the neutrophil (NΦ) is of special interest as this cell is able to uptake nanoparticles (NPs) and cross the vascular barrier in response to tumor signaling. There is a growing body of literature describing NP-NΦ interactions in vitro and in vivo that demonstrates the opportunity of using these cells to improve the efficacy of cancer therapy. However, a number of conceptual and technical issues need to be resolved for translating the technology into clinics. The current review summarizes the recent advances and challenges associated with NP-NΦ interactions, with the special focus on the complex interplay between the NP internalization pathways and the modulation of NΦ activity, and its potential consequences for nanodrug delivery.
Collapse
Affiliation(s)
- Daniil A. Vishnevskiy
- V. Serbsky National Medical Research Center for Psychiatry and Narcology Kropotkinskiy Pereulok, 23 Moscow 119034 Russia
- N. I Pirogov Russian National Research Medical University Ulitsa Ostrovityanova, 1 Moscow 117997 Russia
| | - Anastasiia S. Garanina
- National University of Science and Technology (MISIS) Leninskiy Prospekt, 4 Moscow 119049 Russia
| | - Anastasia A. Chernysheva
- V. Serbsky National Medical Research Center for Psychiatry and Narcology Kropotkinskiy Pereulok, 23 Moscow 119034 Russia
| | - Vladimir P. Chekhonin
- V. Serbsky National Medical Research Center for Psychiatry and Narcology Kropotkinskiy Pereulok, 23 Moscow 119034 Russia
- N. I Pirogov Russian National Research Medical University Ulitsa Ostrovityanova, 1 Moscow 117997 Russia
| | - Victor A. Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology Kropotkinskiy Pereulok, 23 Moscow 119034 Russia
| |
Collapse
|
12
|
Inoue M, Enomoto M, Yoshimura M, Mizowaki T. Pharmacological inhibition of sodium-calcium exchange activates NADPH oxidase and induces infection-independent NETotic cell death. Redox Biol 2021; 43:101983. [PMID: 33933883 PMCID: PMC8105669 DOI: 10.1016/j.redox.2021.101983] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
In addition to its function of innate immunity against invading pathogens, neutrophil extracellular traps (NETs) promote thrombosis, autoimmune disease, and cancer metastasis; therefore, unnecessary exposure to the triggers of infection-independent NET generation should be avoided. We herein show that inhibition of forward-mode Na+/Ca2+ exchange by amiloride analogs, 5-(N-ethyl-N-isopropyl)amiloride (EIPA) and 5-(N-Methyl-N-isobutyl)amiloride (MIA), triggers NETotic cell death independently of infectious stimuli. Isolated human neutrophils treated with EIPA and MIA undergo NETotic cell death by an increase of intracellular Ca2+ following activation of NADPH oxidase and the resultant upregulation of intracellular ROS. EIPA- and MIA-mediated intracellular Ca2+ increase is attributed to the competitive binding of EIPA and MIA against Na+ to Na+/Ca2+ exchanger 1 (NCX1). These results demonstrate a new mechanism of infection-independent NET generation and implicate NCX1 as a physiologic regulator of intracellular calcium balance and NETotic cell death. Two of the amiloride analogs, EIPA and MIA, induce NETotic cell death without infectious stimuli. EIPA and MIA inhibit the forward-mode Na+/Ca2+ exchange and promote the intracellular Ca2+ overload. Intracellular Ca2+ overload by EIPA and MIA activates NADPH oxidase, elevates intracellular ROS level, and induces resultant NETotic cell death.
Collapse
Affiliation(s)
- Minoru Inoue
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Masahiro Enomoto
- Princess Margaret Cancer Centre, Department of Medical Biophysics, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
13
|
Kondo-Kawai A, Sakai T, Terao J, Mukai R. Suppressive effects of quercetin on hydrogen peroxide-induced caveolin-1 phosphorylation in endothelial cells. J Clin Biochem Nutr 2021; 69:28-36. [PMID: 34376911 PMCID: PMC8325769 DOI: 10.3164/jcbn.20-190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/19/2020] [Indexed: 12/29/2022] Open
Abstract
Caveolin-1 is a major protein of the caveolae structure in vascular endothelial cell membrane. Phosphorylation of caveolin-1 is one of the initial events leading to exacerbation of vascular permeability caused by oxidative stress. Although quercetin is known to be an anti-atherosclerosis factor that acts as a dietary antioxidant, little is known about its role in the regulation of caveolin-1 phosphorylation. In this study, we investigated the inhibitory effect of quercetin on hydrogen peroxide-induced caveolin-1 phosphorylation in human umbilical vein endothelial cells. Quercetin inhibited caveolin-1 phosphorylation in cells pretreated with quercetin for 24 h and then exposed to hydrogen peroxide. However, quercetin 3-O-β-glucuronide, a conjugated metabolite of quercetin, did not exert this inhibitory effect. Exposure to hydrogen peroxide increased vascular permeability and reduced mRNA expression of the intercellular adhesion protein, vascular endothelial cadherin (VE-cadherin). By contrast, pretreatment with quercetin suppressed the increase in vascular permeability and decreased VE-cadherin expression. These results indicate that deconjugated quercetin can play a role in the prevention of altered vascular permeability under oxidative stress by suppressing caveolin-1 phosphorylation. Thus, dietary quercetin may be beneficial for the maintenance of endothelial cell function.
Collapse
Affiliation(s)
- Akari Kondo-Kawai
- Department of Food Science, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8513, Japan.,Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima 770-8503, Japan.,Laboratory of Nutritional Science, Shikoku Junior College, Tokushima, Tokushima 771-1192, Japan
| | - Tohru Sakai
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Tokushima 770-8503, Japan
| | - Junji Terao
- Department of Food Science, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8513, Japan.,Faculty of Clinical Nutrition and Dietetics, Konan Women's University, Kobe, Hyogo 658-0001, Japan
| | - Rie Mukai
- Department of Food Science, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8513, Japan.,Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Tokushima 770-8513, Japan
| |
Collapse
|
14
|
Chi Y, Liu X, Chai J. A narrative review of changes in microvascular permeability after burn. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:719. [PMID: 33987417 PMCID: PMC8106041 DOI: 10.21037/atm-21-1267] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/17/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE We aimed to review and discuss some of the latest research results related to post-burn pathophysiological changes and provide some clues for future study. BACKGROUND Burns are one of the most common and serious traumas and consist of a series of pathophysiological changes of thermal injury. Accompanied by thermal damage to skin and soft tissues, inflammatory mediators are released in large quantities. Changes in histamine, bradykinin, and cytokines such as vascular endothelial growth factor (VEGF), metabolic factors such as adenosine triphosphate (ATP), and activated neutrophils all affect the body's vascular permeability. METHODS We searched articles with subject words "microvascular permeability", "burn" "endothelium", and "endothelial barrier" in PubMed in English published from the beginning of database to Dec, 2020. CONCLUSIONS The essence of burn shock is the rapid and extensive fluid transfer in burn and non-burn tissue. After severe burns, the local and systemic vascular permeability increase, causing intravascular fluid extravasation, leading to a progressive decrease in effective circulation volume, an increase in systemic vascular resistance, a decrease in cardiac output, peripheral tissue edema, multiple organ failure, and even death. There are many cells, tissues, mediators and structures involved in the pathophysiological process of the damage to vascular permeability. Ulinastatin is a promising agent for this problem.
Collapse
Affiliation(s)
- Yunfei Chi
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| | - Xiangyu Liu
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| | - Jiake Chai
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Morsing SKH, Rademakers T, Brouns SLN, van Stalborch AMD, Donners MMPC, van Buul JD. ADAM10-Mediated Cleavage of ICAM-1 Is Involved in Neutrophil Transendothelial Migration. Cells 2021; 10:cells10020232. [PMID: 33504031 PMCID: PMC7911467 DOI: 10.3390/cells10020232] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 01/24/2023] Open
Abstract
To efficiently cross the endothelial barrier during inflammation, neutrophils first firmly adhere to the endothelial surface using the endothelial adhesion molecule ICAM-1. Upon actual transmigration, the release from ICAM-1 is required. While Integrin LFA1/Mac1 de-activation is one described mechanism that leads to this, direct cleavage of ICAM-1 from the endothelium represents a second option. We found that a disintegrin and metalloprotease 10 (ADAM10) cleaves the extracellular domain of ICAM-1 from the endothelial surface. Silencing or inhibiting endothelial ADAM10 impaired the efficiency of neutrophils to cross the endothelium, suggesting that neutrophils use endothelial ADAM10 to dissociate from ICAM-1. Indeed, when measuring transmigration kinetics, neutrophils took almost twice as much time to finish the diapedesis step when ADAM10 was silenced. Importantly, we found increased levels of ICAM-1 on the transmigrating neutrophils when crossing an endothelial monolayer where such increased levels were not detected when neutrophils crossed bare filters. Using ICAM-1-GFP-expressing endothelial cells, we show that ICAM-1 presence on the neutrophils can also occur by membrane transfer from the endothelium to the neutrophil. Based on these findings, we conclude that endothelial ADAM10 contributes in part to neutrophil transendothelial migration by cleaving ICAM-1, thereby supporting the release of neutrophils from the endothelium during the final diapedesis step.
Collapse
Affiliation(s)
- Sofia K. H. Morsing
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
| | - Timo Rademakers
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
| | - Sanne L. N. Brouns
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
| | - Anne-Marieke D. van Stalborch
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
| | - Marjo M. P. C. Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
- Correspondence: (M.M.P.C.D.); (J.D.v.B.); Tel.: +31-43-3877167 (M.M.P.C.D.); +31-20-5121219 (J.D.v.B.); Fax: +31-20-5123310 (J.D.v.B.)
| | - Jaap D. van Buul
- Molecular Cell Biology Lab, Department Molecular and Cellular Homeostasis, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (S.K.H.M.); (T.R.); (S.L.N.B.); (A.-M.D.v.S.)
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), Section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS), University of Amsterdam, 1066 CX Amsterdam, The Netherlands
- Correspondence: (M.M.P.C.D.); (J.D.v.B.); Tel.: +31-43-3877167 (M.M.P.C.D.); +31-20-5121219 (J.D.v.B.); Fax: +31-20-5123310 (J.D.v.B.)
| |
Collapse
|
16
|
Cao Y, Chen X, Liu Y, Zhang X, Zou Y, Li J. PIM1 inhibition attenuated endotoxin-induced acute lung injury through modulating ELK3/ICAM1 axis on pulmonary microvascular endothelial cells. Inflamm Res 2021; 70:89-98. [PMID: 33185705 DOI: 10.1007/s00011-020-01420-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 09/26/2020] [Accepted: 11/01/2020] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE The dysfunction of pulmonary microvascular endothelial cells (PMVECs) is one of the critical characteristics of acute lung injury/acute respiratory distress syndrome (ALI/ARDS) induced by severe infection. PIM1 is a constitutively active serine/threonine kinase that is involved in multiple biological processes. However, the underlying correlation between PIM1 and PMVECs injury remains unclear. The main purpose of this study was to reveal roles of PIM1 and explore the potential mechanisms during the development of endotoxin-induced ALI induced by intraperitoneal LPS administration. MATERIALS AND METHODS PIM1 level in the lung tissues of endotoxin-induced ALI mice or plasma derived from cardiopulmonary bypass (CPB)-induced ALI patients were measured. The protective roles of PIM1 specific inhibitor SMI-4a on endotoxin-induced lung injuries were evaluated through histological, permeability, neutrophil infiltration and survival assessment. The relationship between PIM1 and ELK3/ICAM-1 axis was validated in vivo and vitro. The correlation between plasma PIM1 and indicative vascular endothelium injury biomarkers (PaO2/FiO2 ratio, Ang-II, E-selectin and PAI-1) levels derived from CPB-induced ALI patient were analyzed. RESULTS PIM1 expression in the lung tissues was increased in the mice of endotoxin-induced ALI. The PIM1 specific inhibitor SMI-4a administration relieved the severity of endotoxin-induced ALI. More importantly, PIM1 modulates ICAM1 expression through regulating transcription factor ELK3 expression in vitro. Eventually, plasma PIM1 level was positively correlated with Ang-II and PAI-1 levels but negatively correlated with SpO2/FiO2 ratio among CPB induced ALI patients. CONCLUSION Our results indicated that PIM1 inhibition carried a protective role against endotoxin-induced ALI by modulating the ELK3/ICAM1 axis on PMVECs. PIM1 may be a potential therapeutic target for endotoxin-induced ALI.
Collapse
Affiliation(s)
- Yumeng Cao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Xia Chen
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Xingyi Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Yun Zou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China.
| | - Jinbao Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China.
| |
Collapse
|
17
|
Abstract
Transcytosis of macromolecules through lung endothelial cells is the primary route of transport from the vascular compartment into the interstitial space. Endothelial transcytosis is mostly a caveolae-dependent process that combines receptor-mediated endocytosis, vesicle trafficking via actin-cytoskeletal remodeling, and SNARE protein directed vesicle fusion and exocytosis. Herein, we review the current literature on caveolae-mediated endocytosis, the role of actin cytoskeleton in caveolae stabilization at the plasma membrane, actin remodeling during vesicle trafficking, and exocytosis of caveolar vesicles. Next, we provide a concise summary of experimental methods employed to assess transcytosis. Finally, we review evidence that transcytosis contributes to the pathogenesis of acute lung injury. © 2020 American Physiological Society. Compr Physiol 10:491-508, 2020.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard D. Minshall
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Department of Anesthesiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Correspondence to
| |
Collapse
|
18
|
Yang Z, Wang L, Yu H, Wang R, Gou Y, Zhang M, Kang C, Liu T, Lan Y, Wang X, Liu J, Cooper MA, Li X, Yue K, Yu Y, Wang L, Kim BY, Jiang W, Sun W. Membrane TLR9 Positive Neutrophil Mediated MPLA Protects Against Fatal Bacterial Sepsis. Am J Cancer Res 2019; 9:6269-6283. [PMID: 31534550 PMCID: PMC6735515 DOI: 10.7150/thno.37139] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
Sepsis is a major cause of patient mortality and morbidity from bacterial infections. Although neutrophils are known to be important in the development of sepsis, how distinctive neutrophil subtypes regulate inflammatory processes involved in septicemia remains unclear. Preconditioning protects organisms against subsequent higher-dose exposures to the same, or even different, stimuli. Several studies have reported various effects of preconditioning on immune cells. However, the detailed mechanisms underlying neutrophil-mediated protection through preconditioning in sepsis remain unknown. Methods: Flow cytometry was conducted to sort the mice peritoneal lavage cells and the blood samples from patients with sepsis. Western blotting and ELISA were carried out to elucidate the expression of TLR9 signal transduction pathway proteins. Histological analysis was used to assess the effect of InP on intestine and liver structure in tlr9-/- and cav-1-/- mice. Fluorescence microscopy, Co-IP, and FRET were carried out to determine the association of TLR9 with Cav-1. Results: We show that membrane toll-like receptor-9 positive (mTLR9+) neutrophils exert a protective effect against fatal bacterial infections through the process of inflammatory preconditioning (InP). InP, which occurs in the setting of a low-dose bacterial challenge, active ingredient is Monophosphoryl lipid A (MPLA), triggers the membrane translocation of TLR9 from the neutrophil cytosol, where it binds to Cav-1. Our findings showed that InP enables TLR9 to facilitate MyD88-mediated TRAF3 and IRF3 signal transduction. Depletion of either TLR9 or Cav-1 largely eliminates the neutrophil-mediated InP effect in sepsis models in vitro and in vivo. Further, examination of clinical samples from patients with sepsis showed that clinical outcomes and likelihood of recovery are closely correlated with mTLR9 and Cav-1 expression in circulating neutrophils. Conclusion: These results demonstrate that the TLR9-Cav-1 axis is a critical signaling pathway involved in the regulation of neutrophil-dependent MPLA mediated InP, and the presence of mTLR9+ neutrophils could be an attractive indicator of clinical outcomes in bacterial sepsis that could be further explored as a potential therapeutic target.
Collapse
|
19
|
Ma Y, Yang X, Chatterjee V, Meegan JE, Beard Jr. RS, Yuan SY. Role of Neutrophil Extracellular Traps and Vesicles in Regulating Vascular Endothelial Permeability. Front Immunol 2019; 10:1037. [PMID: 31143182 PMCID: PMC6520655 DOI: 10.3389/fimmu.2019.01037] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
The microvascular endothelium serves as the major barrier that controls the transport of blood constituents across the vessel wall. Barrier leakage occurs during infection or sterile inflammation, allowing plasma fluid and cells to extravasate and accumulate in surrounding tissues, an important pathology underlying a variety of infectious diseases and immune disorders. The leak process is triggered and regulated by bidirectional communications between circulating cells and vascular cells at the blood-vessel interface. While the molecular mechanisms underlying this complex process remain incompletely understood, emerging evidence supports the roles of neutrophil-endothelium interaction and neutrophil-derived products, including neutrophil extracellular traps and vesicles, in the pathogenesis of vascular barrier injury. In this review, we summarize the current knowledge on neutrophil-induced changes in endothelial barrier structures, with a detailed presentation of recently characterized molecular pathways involved in the production and effects of neutrophil extracellular traps and extracellular vesicles. Additionally, we discuss the therapeutic implications of altering neutrophil interactions with the endothelial barrier in treating inflammatory diseases.
Collapse
Affiliation(s)
- Yonggang Ma
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Victor Chatterjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jamie E. Meegan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Richard S. Beard Jr.
- Department of Biological Sciences, Biomolecular Research Center, Boise State University, Boise, ID, United States
| | - Sarah Y. Yuan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
20
|
Klein RS, Garber C, Funk KE, Salimi H, Soung A, Kanmogne M, Manivasagam S, Agner S, Cain M. Neuroinflammation During RNA Viral Infections. Annu Rev Immunol 2019; 37:73-95. [PMID: 31026414 PMCID: PMC6731125 DOI: 10.1146/annurev-immunol-042718-041417] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neurotropic RNA viruses continue to emerge and are increasingly linked to diseases of the central nervous system (CNS) despite viral clearance. Indeed, the overall mortality of viral encephalitis in immunocompetent individuals is low, suggesting efficient mechanisms of virologic control within the CNS. Both immune and neural cells participate in this process, which requires extensive innate immune signaling between resident and infiltrating cells, including microglia and monocytes, that regulate the effector functions of antiviral T and B cells as they gain access to CNS compartments. While these interactions promote viral clearance via mainly neuroprotective mechanisms, they may also promote neuropathology and, in some cases, induce persistent alterations in CNS physiology and function that manifest as neurologic and psychiatric diseases. This review discusses mechanisms of RNA virus clearance and neurotoxicity during viral encephalitis with a focus on the cytokines essential for immune and neural cell inflammatory responses and interactions. Understanding neuroimmune communications in the setting of viral infections is essential for the development of treatments that augment neuroprotective processes while limiting ongoing immunopathological processes that cause ongoing CNS disease.
Collapse
Affiliation(s)
- Robyn S Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Charise Garber
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Kristen E Funk
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Hamid Salimi
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Allison Soung
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Marlene Kanmogne
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Sindhu Manivasagam
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| | - Shannon Agner
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Matthew Cain
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA;
| |
Collapse
|
21
|
Abstract
Calpain is an intracellular Ca2+-dependent non-lysosomal cysteine protease expressed ubiquitously in mammals. In endothelial cells, dysregulation of calpain has been shown to be involved in a wide variety of pathological conditions such as angiogenesis, vascular inflammation, and diabetes. Cell- or tissue-targeted in vivo delivery of small interfering RNA (siRNA) is a powerful research tool in the analysis of protein function and has been proposed as an attractive therapeutic modality that is applicable against a large number of human diseases including cancer. In this chapter we describe a method to knockdown calpain 1 in mouse pulmonary vascular endothelium using delivery of siRNA/cationic liposome complex. This technique results in a greater than 80% reduction in calpain 1 protein levels 48 h after a single i.v. injection of calpain 1 siRNA (0.5 mg siRNA/kg)/cationic liposome complex. We also describe confocal imaging to verify the loss of calpain 1 expression in pulmonary microvessel endothelial cells and application of this technique in the mouse model of ventilator-induced lung injury.
Collapse
Affiliation(s)
- Xiaoqian Liu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL, USA.
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL, USA.
| |
Collapse
|
22
|
Simmons S, Erfinanda L, Bartz C, Kuebler WM. Novel mechanisms regulating endothelial barrier function in the pulmonary microcirculation. J Physiol 2018; 597:997-1021. [PMID: 30015354 DOI: 10.1113/jp276245] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary epithelial and vascular endothelial cell layers provide two sequential physical and immunological barriers that together form a semi-permeable interface and prevent alveolar and interstitial oedema formation. In this review, we focus specifically on the continuous endothelium of the pulmonary microvascular bed that warrants strict control of the exchange of gases, fluid, solutes and circulating cells between the plasma and the interstitial space. The present review provides an overview of emerging molecular mechanisms that permit constant transcellular exchange between the vascular and interstitial compartment, and cause, prevent or reverse lung endothelial barrier failure under experimental conditions, yet with a clinical perspective. Based on recent findings and at times seemingly conflicting results we discuss emerging paradigms of permeability regulation by altered ion transport as well as shifts in the homeostasis of sphingolipids, angiopoietins and prostaglandins.
Collapse
Affiliation(s)
- Szandor Simmons
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lasti Erfinanda
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bartz
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Chen Z, D. S. Oliveira S, Zimnicka AM, Jiang Y, Sharma T, Chen S, Lazarov O, Bonini MG, Haus JM, Minshall RD. Reciprocal regulation of eNOS and caveolin-1 functions in endothelial cells. Mol Biol Cell 2018; 29:1190-1202. [PMID: 29563255 PMCID: PMC5935069 DOI: 10.1091/mbc.e17-01-0049] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/12/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
We hypothesized that the maintenance of vascular homeostasis is critically dependent on the expression and reciprocal regulation of caveolin-1 (Cav-1) and endothelial nitric oxide synthase (eNOS) in endothelial cells (ECs). Skeletal muscle biopsies from subjects with type 2 diabetes showed 50% less Cav-1 and eNOS than those from lean healthy controls. The Cav-1:eNOS expression ratio was 200:1 in primary culture human ECs. Cav-1 small interfering RNA (siRNA) reduced eNOS protein and gene expression in association with a twofold increase in eNOS phosphorylation and nitrate production per molecule of eNOS, which was reversed in cells overexpressing Adv-Cav-1-GFP. Upon addition of the Ca2+ ionophore A23187 to activate eNOS, we observed eNOS Ser1177 phosphorylation, its translocation to β-catenin-positive cell-cell junctions, and increased colocalization of eNOS and Cav-1 within 5 min. We also observed Cav-1 S-nitrosylation and destabilization of Cav-1 oligomers in cells treated with A23187 as well as insulin or albumin, and this could be blocked by L-NAME, PP2, or eNOS siRNA. Finally, caveola-mediated endocytosis of albumin or insulin was reduced by Cav-1 or eNOS siRNA, and the effect of Cav-1 siRNA was rescued by Adv-Cav-1-GFP. Thus, Cav-1 stabilizes eNOS expression and regulates its activity, whereas eNOS-derived NO promotes caveola-mediated endocytosis.
Collapse
Affiliation(s)
- Zhenlong Chen
- Departments of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612
| | | | | | - Ying Jiang
- Departments of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612
| | - Tiffany Sharma
- Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Stone Chen
- Whitney M. Young Magnet High School, Chicago, IL 60607
| | - Orly Lazarov
- Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Jacob M. Haus
- Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Richard D. Minshall
- Departments of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612
- Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| |
Collapse
|
24
|
Transcytosis Involvement in Transport System and Endothelial Permeability of Vascular Leakage during Dengue Virus Infection. Viruses 2018; 10:v10020069. [PMID: 29419739 PMCID: PMC5850376 DOI: 10.3390/v10020069] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/19/2022] Open
Abstract
The major role of endothelial cells is to maintain homeostasis of vascular permeability and to preserve the integrity of vascular vessels to prevent fluid leakage. Properly functioning endothelial cells promote physiological balance and stability for blood circulation and fluid components. A monolayer of endothelial cells has the ability to regulate paracellular and transcellular pathways for transport proteins, solutes, and fluid. In addition to the paracellular pathway, the transcellular pathway is another route of endothelial permeability that mediates vascular permeability under physiologic conditions. The transcellular pathway was found to be associated with an assortment of disease pathogeneses. The clinical manifestation of severe dengue infection in humans is vascular leakage and hemorrhagic diatheses. This review explores and describes the transcellular pathway, which is an alternate route of vascular permeability during dengue infection that corresponds with the pathologic finding of intact tight junction. This pathway may be the route of albumin transport that causes endothelial dysfunction during dengue virus infection.
Collapse
|
25
|
Chamaraux-Tran TN, Piegeler T. The Amide Local Anesthetic Lidocaine in Cancer Surgery-Potential Antimetastatic Effects and Preservation of Immune Cell Function? A Narrative Review. Front Med (Lausanne) 2017; 4:235. [PMID: 29326939 PMCID: PMC5742360 DOI: 10.3389/fmed.2017.00235] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/06/2017] [Indexed: 12/13/2022] Open
Abstract
Surgical removal of the primary tumor in solid cancer is an essential component of the treatment. However, the perioperative period can paradoxically lead to an increased risk of cancer recurrence. A bimodal dynamics for early-stage breast cancer recurrence suggests a tumor dormancy-based model with a mastectomy-driven acceleration of the metastatic process and a crucial role of the immunosuppressive state during the perioperative period. Recent evidence suggests that anesthesia could also influence the progress of the disease. Local anesthetics (LAs) have long been used for their properties to block nociceptive input. They also exert anti-inflammatory capacities by modulating the liberation or signal propagation of inflammatory mediators. Interestingly, LAs can reduce viability and proliferation of many cancer cells in vitro as well. Additionally, retrospective clinical trials have suggested that regional anesthesia for cancer surgery (either with or without general anesthesia) might reduce the risk of recurrence. Lidocaine, a LA, which can be administered intravenously, is widely used in clinical practice for multimodal analgesia. It is associated with a morphine-sparing effect, reduced pain scores, and in major surgery probably also with a reduced incidence of postoperative ileus and length of hospital stay. Systemic delivery might therefore be efficient to target residual disease or reach cells able to form micrometastasis. Moreover, an in vitro study has shown that lidocaine could enhance the activity of natural killer (NK) cells. Due to their ability to recognize and kill tumor cells without the requirement of prior antigen exposure, NKs are the main actor of the innate immune system. However, several perioperative factors can reduce NK activity, such as stress, pain, opioids, or general anesthetics. Intravenous lidocaine as part of the perioperative anesthesia regimen would be of major interest for clinicians, as it might bear the potential to reduce the risk of cancer recurrence or progression patients undergoing cancer surgery. As a well-known pharmaceutical agent, lidocaine might therefore be a promising candidate for oncological drug repurposing. We urgently need clinical randomized trials assessing the protective effect of lidocaine on NKs function and against recurrence after cancer surgery to achieve a “proof of concept.”
Collapse
Affiliation(s)
- Thiên-Nga Chamaraux-Tran
- Département d'Anesthésie et Réanimation Chirurgicale, Hôpital Hautepierre, CHU Strasbourg, Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, Unité Mixte de Recherche 7104, Centre National de la Recherche Scientifique, U964 Institut National de Santé et de Recherche Médicale, Université de Strasbourg, Illkirch, France
| | - Tobias Piegeler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
26
|
ICAM-1 regulates macrophage polarization by suppressing MCP-1 expression via miR-124 upregulation. Oncotarget 2017; 8:111882-111901. [PMID: 29340098 PMCID: PMC5762366 DOI: 10.18632/oncotarget.22948] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022] Open
Abstract
Intercellular adhesion molecule-1 is the adhesion molecule mediating leukocyte firm adhesion to endothelial cells, plays a critical role in subsequent leukocyte transmigration. ICAM-1 is also expressed in other cells including macrophages; however, the role of this adhesion molecule in mediating macrophage functions remains enigmatic. We report that ICAM-1 regulates macrophage polarization by positively modulating miR-124 expression. We found higher expression levels of monocyte chemotactic protein-1 in lungs of mice lacking ICAM-1. Consistent with this result, siRNA mediated depletion of ICAM-1 in macrophage resulted in increased expression levels of MCP-1. Moreover, ICAM-1 controlled miR-124 expression and downregulated MCP-1 mRNA and protein expression by binding of miR-124 to MCP-1 3' untranslated region. ICAM-1 also induced the transcription factor Sp1 expression, which is important for miR-124 expressing in macrophages. Furthermore, ICAM-1 depletion led to M1 macrophage polarization, in contrast, miR-124 mimics promoted M2 macrophage polarization. Exogenous administration of miR-124 mimics into the lungs prevented lipopolysaccharide-induced myeloperoxidase activity in vivo, suggesting that miR-124 is important for dampening acute lung injury. These results collectively show that adhesion molecule ICAM-1 downregulates MCP-1 expression by controlling Sp1 mediated miR-124 levels, which in turn regulate M2 macrophage polarization. Targeting ICAM-1 and downstream miR-124 may present a new therapeutic strategy for acute lung injury.
Collapse
|
27
|
Ono S, Egawa G, Kabashima K. Regulation of blood vascular permeability in the skin. Inflamm Regen 2017; 37:11. [PMID: 29259710 PMCID: PMC5725833 DOI: 10.1186/s41232-017-0042-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/25/2017] [Indexed: 12/31/2022] Open
Abstract
Regulation of blood vessel permeability is essential for the homeostasis of peripheral tissues. This regulation controls the trafficking of plasma contents, including water, vitamins, ions, hormones, cytokines, amyloids, lipoproteins, carrier proteins, and immunoglobulins. The properties of blood vessels vary among tissues based on their structural differences: continuous, fenestrated, or sinusoidal. These three types of blood vessels have different charge and size barrier properties. The anionic luminal glycocalyx layer on endothelial cells establishes the "charge barrier" that repels the attachment of negatively charged blood cells and plasma molecules. In contrast, the "size barrier" of blood vessels largely relies on the interendothelial junctions (IEJs) between endothelial cells, which define the paracellular permeability. As in most peripheral tissues, blood capillaries in the skin are composed of continuous and/or fenestrated blood vessels that have relatively tighter IEJs compared to those in the internal organs. Small vesicles in the capillary endothelium were discovered in the 1950s, and studies have since confirmed that blood endothelial cells transport the plasma contents by endocytosis and subsequent transcytosis and exocytosis-this process is called transcellular permeability. The permeability of blood vessels is highly variable as a result of intrinsic and extrinsic factors. It is significantly elevated upon tissue inflammations as a result of disabled IEJs and increased paracellular permeability due to inflammatory mediators. An increase in transcellular permeability during inflammation has also been postulated. Here, we provide an overview of the general properties of vascular permeability based on our recent observations of murine skin inflammation models, and we discuss its physiological significance in peripheral homeostasis.
Collapse
Affiliation(s)
- Sachiko Ono
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara, Sakyo, Kyoto, 606-8507 Japan
| | - Gyohei Egawa
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara, Sakyo, Kyoto, 606-8507 Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara, Sakyo, Kyoto, 606-8507 Japan
- Singapore Immunology Network (SIgN) and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
- PRESTO, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
28
|
Increased Extravascular Lung Water and Plasma Biomarkers of Acute Lung Injury Precede Oxygenation Impairment in Primary Graft Dysfunction After Lung Transplantation. Transplantation 2017; 101:112-121. [PMID: 27495752 DOI: 10.1097/tp.0000000000001434] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND After lung transplantation (LT), early prediction of grade 3 pulmonary graft dysfunction (PGD) remains a research gap for clinicians. We hypothesized that it could be improved using extravascular lung water (EVLWi) and plasma biomarkers of acute lung injury. METHODS After institutional review board approval and informed consent, consecutive LT recipients were included. Transpulmonary thermodilution-based EVLWi, plasma concentrations of epithelial (soluble receptor for advanced glycation endproducts [sRAGE]) and endothelial biomarkers (soluble intercellular adhesion molecule-1 and endocan [full-length and cleaved p14 fragment]) were obtained before and after LT (0 [H0], 6, 12, 24, 48 and 72 hours after pulmonary artery unclamping). Grade 3 PGD was defined according to the International Society for Lung and Heart Transplantation definition, combining arterial oxygen partial pressure (PaO2)/inspired fraction of oxygen (FiO2) ratio and chest X-rays. Association of clinical risk factors, EVLWi and biomarkers with grade 3 PGD was analyzed under the Bayesian paradigm, using logistic model and areas under the receiver operating characteristic curves (AUCs). RESULTS In 47 LT recipients, 10 developed grade 3 PGD, which was obvious at H6 in 8 cases. Clinical risk factors, soluble intercellular adhesion molecule-1 and endocan (both forms) were not associated with grade 3 PGD. Significant predictors of grade 3 PGD included (1) EVLWi (optimal cutoff, 13.7 mL/kg; AUC, 0.74; 95% confidence interval [CI], 0.48-0.99), (2) PaO2/FiO2 ratio (optimal cutoff, 236; AUC, 0.68; 95% CI, 0.52-0.84), and (3) sRAGE (optimal cutoff, 11 760 pg/mL; AUC, 0.66; 95% CI, 0.41-0.91) measured at H0. CONCLUSIONS Immediate postreperfusion increases in EVLWi and sRAGE along with impaired PaO2/FiO2 ratios were early predictors of grade 3 PGD at or beyond 6 hours and may trigger early therapeutic interventions.
Collapse
|
29
|
Xiao MM, Pan CS, Liu YY, Ma LQ, Yan L, Fan JY, Wang CS, Huang R, Han JY. Post-treatment with Ma-Huang-Tang ameliorates cold-warm-cycles induced rat lung injury. Sci Rep 2017; 7:312. [PMID: 28331194 PMCID: PMC5428516 DOI: 10.1038/s41598-017-00459-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/27/2017] [Indexed: 01/22/2023] Open
Abstract
Frequent and drastic ambient temperature variation may cause respiratory diseases such as common cold and pneumonia, the mechanism for which is not fully understood, however, due to lack of appropriate animal models. Ma-Huang-Tang (MHT) is widely used in China for treatment of respiratory diseases. The present study aimed to investigate the effect of MHT on temperature alternation induced rat lung injury and explore underlying mechanisms. Male Sprague-Dawley rats were exposed to a cold environment for 1 h and then shifted to a warm environment for 30 min. This cold and warm alteration cycled 4 times. Rats were administrated with MHT (1.87 g/kg) by gavage 6 h after cold-warm-cycles. Cold-warm-cycles induced pulmonary microcirculatory disorders, lung edema and injury, decrease in the expression of tight junction proteins, increase in VE-cadherin activation, increase in the expression and activation of Caveolin-1, Src and NF-κB, and NADPH oxidase subunits p47phox, p40phox and p67phox membrane translocation and inflammatory cytokines production. All alterations were significantly ameliorated by post-treatment with MHT. This study showed that rats subjected to cold-warm-cycles may be used as an animal model to investigate ambient temperature variation-induced lung injury, and suggested MHT as a potential strategy to combat lung injury induced by temperature variation.
Collapse
Affiliation(s)
- Meng-Meng Xiao
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China
| | - Yu-Ying Liu
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China
| | - Li-Qian Ma
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China
| | - Li Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China
| | - Chuan-She Wang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China
| | - Rong Huang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China. .,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, China. .,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China. .,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, 100191, China.
| |
Collapse
|
30
|
Huang Y, He Q. Inhibition of c-Src protects paraquat induced microvascular endothelial injury by modulating caveolin-1 phosphorylation and caveolae mediated transcellular permeability. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 52:62-68. [PMID: 28376378 DOI: 10.1016/j.etap.2017.01.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 02/05/2023]
Abstract
The mechanisms underlying paraquat induced acute lung injury (ALI) is still not clear. C-Src plays an important role in the regulation of microvascular endothelial barrier function and the pathogenesis of ALI. In the present study, we found that paraquat induced cell toxicity and an increase of reactive oxygen species (ROS) in endothelium. Paraquat exposure also induced significant increase of caveolin-1 phosphorylation, caveolae trafficking and albumin permeability in endothelial monolayers. C-Src depletion by siRNA significantly attenuate paraquat induced cell toxicity, caveolin-1 phosphorylation, caveolae formation and endothelial hyperpermeability. N-acetylcysteine (NAC) failed to protect endothelial monolayers against paraquat induced toxicity. Thus, our findings suggest that paraquat exposure increases paracellular endothelial permeability by increasing caveolin-1 phosphorylation in a c-Src dependant manner. The depletion of c-Src might protect microvascular endothelial function by regulating caveolin-1 phosphorylation and caveolae trafficking during paraquat exposure, and might have potential therapeutic effects on paraquat induced ALI.
Collapse
Affiliation(s)
- Yu Huang
- Department of Intensive Care Medicine, The Third People's Hospital of Chengdu, The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Qing He
- Department of Intensive Care Medicine, The Third People's Hospital of Chengdu, The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China; Department of Respiratory Disease, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
31
|
Gillrie MR, Ho M. Dynamic interactions of Plasmodium spp. with vascular endothelium. Tissue Barriers 2017; 5:e1268667. [PMID: 28452684 PMCID: PMC5362994 DOI: 10.1080/21688370.2016.1268667] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/24/2016] [Accepted: 11/30/2016] [Indexed: 12/18/2022] Open
Abstract
Plasmodial species are protozoan parasites that infect erythrocytes. As such, they are in close contact with microvascular endothelium for most of the life cycle in the mammalian host. The host-parasite interactions of this stage of the infection are responsible for the clinical manifestations of the disease that range from a mild febrile illness to severe and frequently fatal syndromes such as cerebral malaria and multi-organ failure. Plasmodium falciparum, the causative agent of the most severe form of malaria, is particularly predisposed to modulating endothelial function through either direct adhesion to endothelial receptor molecules, or by releasing potent host and parasite products that can stimulate endothelial activation and/or disrupt barrier function. In this review, we provide a critical analysis of the current clinical and laboratory evidence for endothelial dysfunction during severe P. falciparum malaria. Future investigations using state-of-the-art technologies such as mass cytometry and organs-on-chips to further delineate parasite-endothelial cell interactions are also discussed.
Collapse
Affiliation(s)
- Mark R. Gillrie
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - May Ho
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Coelho-Santos V, Socodato R, Portugal C, Leitão RA, Rito M, Barbosa M, Couraud PO, Romero IA, Weksler B, Minshall RD, Fontes-Ribeiro C, Summavielle T, Relvas JB, Silva AP. Methylphenidate-triggered ROS generation promotes caveolae-mediated transcytosis via Rac1 signaling and c-Src-dependent caveolin-1 phosphorylation in human brain endothelial cells. Cell Mol Life Sci 2016; 73:4701-4716. [PMID: 27376435 PMCID: PMC11108272 DOI: 10.1007/s00018-016-2301-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 06/06/2016] [Accepted: 06/28/2016] [Indexed: 12/14/2022]
Abstract
Methylphenidate (MPH) is an amphetamine-like stimulant commonly prescribed for attention deficit hyperactivity disorder. Despite its widespread use, the cellular/molecular effects of MPH remain elusive. Here, we report a novel direct role of MPH on the regulation of macromolecular flux through human brain endothelial cells (ECs). MPH significantly increased caveolae-mediated transcytosis of horseradish peroxidase through ECs without affecting paracellular permeability. Using FRET-based live cell imaging, together with pharmacological inhibitors and lentiviral-mediated shRNA knockdown, we demonstrate that MPH promoted ROS generation via activation of Rac1-dependent NADPH oxidase (NOX) and c-Src activation at the plasma membrane. c-Src in turn was shown to mediate the phosphorylation of caveolin-1 (Cav1) on Tyr14 leading to enhanced caveolae formation and transendothelial transport. Accordingly, the inhibition of Cav1 phosphorylation by overexpression of a phosphodefective Cav1Y14F mutant or knocking down Cav1 expression abrogated MPH-induced transcytosis. In addition, both vitamin C and inhibition of NOX blocked MPH-triggered vesicular transport. This study, therefore, identifies Rac1/NOX/c-Src-dependent signaling in MPH-induced increase in transendothelial permeability of brain endothelial cell monolayers via caveolae-mediated transcytosis.
Collapse
Affiliation(s)
- Vanessa Coelho-Santos
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Renato Socodato
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Camila Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Ricardo A Leitão
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Manuel Rito
- Neurosurgery Service, Coimbra Hospital and University Centre (CHUC), Coimbra, Portugal
| | - Marcos Barbosa
- Neurosurgery Service, Coimbra Hospital and University Centre (CHUC), Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Ignacio A Romero
- Department of Life Sciences, Faculty of Science, The Open University, Milton Keynes, UK
| | - Babette Weksler
- Department of Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Carlos Fontes-Ribeiro
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Teresa Summavielle
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Ana P Silva
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- CNC.IBILI, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
33
|
Gu X, Reagan AM, McClellan ME, Elliott MH. Caveolins and caveolae in ocular physiology and pathophysiology. Prog Retin Eye Res 2016; 56:84-106. [PMID: 27664379 DOI: 10.1016/j.preteyeres.2016.09.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 09/15/2016] [Accepted: 09/20/2016] [Indexed: 12/14/2022]
Abstract
Caveolae are specialized, invaginated plasma membrane domains that are defined morphologically and by the expression of signature proteins called, caveolins. Caveolae and caveolins are abundant in a variety of cell types including vascular endothelium, glia, and fibroblasts where they play critical roles in transcellular transport, endocytosis, mechanotransduction, cell proliferation, membrane lipid homeostasis, and signal transduction. Given these critical cellular functions, it is surprising that ablation of the caveolae organelle does not result in lethality suggesting instead that caveolae and caveolins play modulatory roles in cellular homeostasis. Caveolar components are also expressed in ocular cell types including retinal vascular cells, Müller glia, retinal pigment epithelium (RPE), conventional aqueous humor outflow cells, the corneal epithelium and endothelium, and the lens epithelium. In the eye, studies of caveolae and other membrane microdomains (i.e., "lipid rafts") have lagged behind what is a substantial body of literature outside vision science. However, interest in caveolae and their molecular components has increased with accumulating evidence of important roles in vision-related functions such as blood-retinal barrier homeostasis, ocular inflammatory signaling, pathogen entry at the ocular surface, and aqueous humor drainage. The recent association of CAV1/2 gene loci with primary open angle glaucoma and intraocular pressure has further enhanced the need to better understand caveolar functions in the context of ocular physiology and disease. Herein, we provide the first comprehensive review of literature on caveolae, caveolins, and other membrane domains in the context of visual system function. This review highlights the importance of caveolae domains and their components in ocular physiology and pathophysiology and emphasizes the need to better understand these important modulators of cellular function.
Collapse
Affiliation(s)
- Xiaowu Gu
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Alaina M Reagan
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mark E McClellan
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael H Elliott
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
34
|
Shang D, Peng T, Gou S, Li Y, Wu H, Wang C, Yang Z. High Mobility Group Box Protein 1 Boosts Endothelial Albumin Transcytosis through the RAGE/Src/Caveolin-1 Pathway. Sci Rep 2016; 6:32180. [PMID: 27572515 PMCID: PMC5004123 DOI: 10.1038/srep32180] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 08/03/2016] [Indexed: 12/25/2022] Open
Abstract
High-mobility group box protein 1 (HMGB1), an inflammatory mediator, has been reported to destroy cell-cell junctions, resulting in vascular endothelial hyperpermeability. Here, we report that HMGB1 increases the endothelial transcytosis of albumin. In mouse lung vascular endothelial cells (MLVECs), HMGB1 at a concentration of 500 ng/ml or less did not harm cell-cell junctions but rapidly induced endothelial hyperpermeability to 125I-albumin. HMGB1 induced an increase in 125I-albumin and AlexaFluor 488-labeled albumin internalization in endocytosis assays. Depletion of receptor for advanced glycation end products (RAGE), but not TLR2 or TLR4, suppressed HMGB1-induced albumin transcytosis and endocytosis. Genetic and pharmacological destruction of lipid rafts significantly inhibited HMGB1-induced albumin endocytosis and transcytosis. HMGB1 induced the rapid phosphorylation of caveolin (Cav)-1 and Src. Either RAGE gene silencing or soluble RAGE suppressed Cav-1 Tyr14 phosphorylation and Src Tyr418 phosphorylation. The Src inhibitor 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo[3,4-d] pyrimidine (PP2) blocked HMGB1-induced Cav-1 Tyr14 phosphorylation. PP2 and overexpression of Cav-1 with a T14F mutation significantly inhibited HMGB1-induced transcytosis and albumin endocytosis. Our findings suggest that HMGB1 induces the transcytosis of albumin via RAGE-dependent Src phosphorylation and Cav-1 phosphorylation. These studies revealed a new mechanism of HMGB1-induced endothelial hyperpermeability.
Collapse
Affiliation(s)
- Dan Shang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Tao Peng
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Shanmiao Gou
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Heshui Wu
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Chunyou Wang
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Zhiyong Yang
- Department of Pancreatic surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| |
Collapse
|
35
|
Caveolae-mediated albumin transcytosis is enhanced in dengue-infected human endothelial cells: A model of vascular leakage in dengue hemorrhagic fever. Sci Rep 2016; 6:31855. [PMID: 27546060 PMCID: PMC4992822 DOI: 10.1038/srep31855] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/28/2016] [Indexed: 01/04/2023] Open
Abstract
Vascular leakage is a life-threatening complication of dengue virus (DENV) infection. Previously, association between “paracellular” endothelial hyperpermeability and plasma leakage had been extensively investigated. However, whether “transcellular” endothelial leakage is involved in dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) remained unknown. We thus investigated effects of DENV (serotype 2) infection on transcellular transport of albumin, the main oncotic plasma protein, through human endothelial cell monolayer by Western blotting, immunofluorescence staining, fluorescence imaging, and fluorometry. The data showed that Alexa488-conjugated bovine serum albumin (Alexa488-BSA) was detectable inside DENV2-infected cells and its level was progressively increased during 48-h post-infection. While paracellular transport could be excluded using FITC-conjugated dextran, Alexa488-BSA was progressively increased and decreased in lower and upper chambers of Transwell, respectively. Pretreatment with nystatin, an inhibitor of caveolae-dependent endocytic pathway, significantly decreased albumin internalization into the DENV2-infected cells, whereas inhibitors of other endocytic pathways showed no significant effects. Co-localization of the internalized Alexa488-BSA and caveolin-1 was also observed. Our findings indicate that DENV infection enhances caveolae-mediated albumin transcytosis through human endothelial cells that may ultimately induce plasma leakage from intravascular compartment. Further elucidation of this model in vivo may lead to effective prevention and better therapeutic outcome of DHF/DSS.
Collapse
|
36
|
Muradashvili N, Tyagi R, Tyagi N, Tyagi SC, Lominadze D. Cerebrovascular disorders caused by hyperfibrinogenaemia. J Physiol 2016; 594:5941-5957. [PMID: 27121987 DOI: 10.1113/jp272558] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/25/2016] [Indexed: 01/18/2023] Open
Abstract
KEY POINTS Hyperfibrinogenaemia (HFg) results in vascular remodelling, and fibrinogen (Fg) and amyloid β (Aβ) complex formation is a hallmark of Alzheimer's disease. However, the interconnection of these effects, their mechanisms and implications in cerebrovascular diseases are not known. Using a mouse model of HFg, we showed that at an elevated blood level, Fg increases cerebrovascular permeability via mainly caveolar protein transcytosis. This enhances deposition of Fg in subendothelial matrix and interstitium making the immobilized Fg a readily accessible substrate for binding Aβ and cellular prion protein (PrPC ), the protein that is thought to have a greater effect on memory than Aβ. We showed that enhanced formation of Fg-Aβ and Fg-PrPC complexes are associated with reduction in short-term memory. The present study delineates a new mechanistic pathway for vasculo-neuronal dysfunctions found in inflammatory cardiovascular and cerebrovascular diseases associated with an elevated blood level of Fg. ABSTRACT Many cardiovascular diseases are associated with inflammation and as such are accompanied by an increased blood level of fibrinogen (Fg). Besides its well-known prothrombotic effects Fg seems to have other destructive roles in developing microvascular dysfunction that include changes in vascular reactivity and permeability. Increased permeability of brain microvessels has the most profound effects as it may lead to cerebrovascular remodelling and result in memory reduction. The goal of the present study was to define mechanisms of cerebrovascular permeability and associated reduction in memory induced by elevated blood content of Fg. Genetically modified, transgenic hyperfibrinogenic (HFg) mice were used to study cerebrovascular transcellular and paracellular permeability in vivo. The extent of caveolar formation and the role of caveolin-1 signalling were evaluated by immunohistochemistry (IHC) and Western blot (WB) analysis in brain samples from experimental animals. Formation of Fg complexes with amyloid β (Aβ) and with cellular prion protein (PrPC ) were also assessed with IHC and WB analysis. Short-term memory of mice was assessed by novel object recognition and Y-maze tests. Caveolar protein transcytosis was found to have a prevailing role in overall increased cerebrovascular permeability in HFg mice. These results were associated with enhanced formation of caveolae. Increased formation of Fg-PrPC and Fg-Aβ complexes were correlated with reduction in short-term memory in HFg mice. Using the model of hyperfibrinogenaemia, the present study shows a novel mechanistic pathway of inflammation-induced and Fg-mediated reduction in short-term memory.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Reeta Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Neetu Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA.
| |
Collapse
|
37
|
Di A, Mehta D, Malik AB. ROS-activated calcium signaling mechanisms regulating endothelial barrier function. Cell Calcium 2016; 60:163-71. [PMID: 26905827 DOI: 10.1016/j.ceca.2016.02.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 02/07/2023]
Abstract
Increased vascular permeability is a common pathogenic feature in many inflammatory diseases. For example in acute lung injury (ALI) and its most severe form, the acute respiratory distress syndrome (ARDS), lung microvessel endothelia lose their junctional integrity resulting in leakiness of the endothelial barrier and accumulation of protein rich edema. Increased reactive oxygen species (ROS) generated by neutrophils (PMNs) and other inflammatory cells play an important role in increasing endothelial permeability. In essence, multiple inflammatory syndromes are caused by dysfunction and compromise of the barrier properties of the endothelium as a consequence of unregulated acute inflammatory response. This review focuses on the role of ROS signaling in controlling endothelial permeability with particular focus on ALI. We summarize below recent progress in defining signaling events leading to increased endothelial permeability and ALI.
Collapse
Affiliation(s)
- Anke Di
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, United States
| | - Dolly Mehta
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, United States
| | - Asrar B Malik
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, United States.
| |
Collapse
|
38
|
Kuebler WM, Wittenberg C, Lee WL, Reppien E, Goldenberg NM, Lindner K, Gao Y, Winoto-Morbach S, Drab M, Mühlfeld C, Dombrowsky H, Ochs M, Schütze S, Uhlig S. Thrombin stimulates albumin transcytosis in lung microvascular endothelial cells via activation of acid sphingomyelinase. Am J Physiol Lung Cell Mol Physiol 2016; 310:L720-32. [PMID: 26851257 DOI: 10.1152/ajplung.00157.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 01/22/2016] [Indexed: 01/12/2023] Open
Abstract
Transcellular albumin transport occurs via caveolae that are abundant in lung microvascular endothelial cells. Stimulation of albumin transcytosis by proinflammatory mediators may contribute to alveolar protein leak in lung injury, yet the regulation of albumin transport and its underlying molecular mechanisms are so far incompletely understood. Here we tested the hypothesis that thrombin may stimulate transcellular albumin transport across lung microvascular endothelial cells in an acid-sphingomyelinase dependent manner. Thrombin increased the transport of fluorescently labeled albumin across confluent human lung microvascular endothelial cell (HMVEC-L) monolayers to an extent that markedly exceeds the rate of passive diffusion. Thrombin activated acid sphingomyelinase (ASM) and increased ceramide production in HMVEC-L, but not in bovine pulmonary artery cells, which showed little albumin transport in response to thrombin. Thrombin increased total caveolin-1 (cav-1) content in both whole cell lysates and lipid rafts from HMVEC-L, and this effect was blocked by inhibition of ASM or de novo protein biosynthesis. Thrombin-induced uptake of albumin into lung microvascular endothelial cells was confirmed in isolated-perfused lungs by real-time fluorescence imaging and electron microscopy of gold-labeled albumin. Inhibition of ASM attenuated thrombin-induced albumin transport both in confluent HMVEC-L and in intact lungs, whereas HMVEC-L treatment with exogenous ASM increased albumin transport and enriched lipid rafts in cav-1. Our findings indicate that thrombin stimulates transcellular albumin transport in an acid sphingomyelinase-dependent manner by inducing de novo synthesis of cav-1 and its recruitment to membrane lipid rafts.
Collapse
Affiliation(s)
- Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Heart Institute Berlin, Berlin, Germany; The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada; Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Claudia Wittenberg
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Warren L Lee
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada; Interdepartmental Division of Critical Care, Departments of Medicine and Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Eike Reppien
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Neil M Goldenberg
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada
| | - Karsten Lindner
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Yizhuo Gao
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada
| | | | - Marek Drab
- Institute of Immunology and Experimental Therapy, Wroclaw, Poland
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DLZ), Hannover, Germany; Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany; and
| | - Heike Dombrowsky
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DLZ), Hannover, Germany; Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany; and
| | - Stefan Schütze
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Stefan Uhlig
- Division of Pulmonary Pharmacology, Research Center Borstel, Borstel, Germany; Institute of Pharmacology and Toxicology, RWTH Aachen, Aachen, Germany
| |
Collapse
|
39
|
Yan Z, Wang ZG, Segev N, Hu S, Minshall RD, Dull RO, Zhang M, Malik AB, Hu G. Rab11a Mediates Vascular Endothelial-Cadherin Recycling and Controls Endothelial Barrier Function. Arterioscler Thromb Vasc Biol 2016; 36:339-349. [PMID: 26663395 PMCID: PMC4732894 DOI: 10.1161/atvbaha.115.306549] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/24/2015] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Vascular endothelial (VE)-cadherin is the predominant component of endothelial adherens junctions essential for cell-cell adhesion and formation of the vascular barrier. Endocytic recycling is an important mechanism for maintaining the expression of cell surface membrane proteins. However, little is known about the molecular mechanism of VE-cadherin recycling and its role in maintenance of vascular integrity. APPROACH AND RESULTS Using calcium-switch assay, confocal imaging, cell surface biotinylation, and flow cytometry, we showed that VE-cadherin recycling required Ras-related proteins in brain (Rab)11a and Rab11 family-interacting protein 2. Yeast 2-hybrid assay and coimmunoprecipitation demonstrated that direct interaction of VE-cadherin with family-interacting protein 2 (at aa 453-484) formed a ternary complex with Rab11a in human endothelial cells. Silencing of Rab11a or Rab11 family-interacting protein 2 in endothelial cells prevented VE-cadherin recycling and VE-cadherin expression at endothelial plasma membrane. Furthermore, inactivation of Rab11a signaling blocked junctional reannealing after vascular inflammation. Selective knockdown of Rab11a in pulmonary microvessels markedly increased vascular leakage in mice challenged with lipopolysaccharide or polymicrobial sepsis. CONCLUSIONS Rab11a/Rab11 family-interacting protein 2-mediated VE-cadherin recycling is required for formation of adherens junctions and restoration of VE barrier integrity and hence a potential target for clinical intervention in inflammatory disease.
Collapse
Affiliation(s)
- Zhibo Yan
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.)
| | - Zhen-Guo Wang
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.)
| | - Nava Segev
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.)
| | - Sanyuan Hu
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.)
| | - Richard D Minshall
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.)
| | - Randal O Dull
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.)
| | - Meihong Zhang
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.)
| | - Asrar B Malik
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.)
| | - Guochang Hu
- From the Departments of Anesthesiology (Z.Y., Z.-G.W., R.D.M., R.O.D., M.Z., G.H.), Pharmacology (Z.Y., R.D.M., A.B.M., G.H.), and Biochemistry and Molecular Genetics (N.S.), University of Illinois College of Medicine, Chicago; and Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China (Z.Y., S.H.).
| |
Collapse
|
40
|
Zhang J, Jiang Z, Bao C, Mei J, Zhu J. Cardiopulmonary bypass increases pulmonary microvascular permeability through the Src kinase pathway: Involvement of caveolin-1 and vascular endothelial cadherin. Mol Med Rep 2016; 13:2918-24. [PMID: 26847917 PMCID: PMC4768976 DOI: 10.3892/mmr.2016.4831] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 01/12/2016] [Indexed: 11/22/2022] Open
Abstract
Changes in pulmonary microvascular permeability following cardiopulmonary bypass (CPB) and the underlying mechanisms have not yet been established. Therefore, the aim of the present study was to elucidate the alterations in pulmonary microvascular permeability following CPB and the underlying mechanism. The pulmonary microvascular permeability was measured using Evans Blue dye (EBD) exclusion, and the neutrophil infiltration and proinflammatory cytokine secretion was investigated. In addition, the activation of Src kinase and the phosphorylation of caveolin-1 and vascular endothelial cadherin (VE-cadherin) was examined. The results revealed that CPB increased pulmonary microvascular leakage, neutrophil count and proinflammatory cytokines in the bronchoalveolar lavage fluid, and activated Src kinase. The administration of PP2, an inhibitor of Src kinase, decreased the activation of Src kinase and attenuated the increase in pulmonary microvascular permeability observed following CPB. Two important proteins associated with vascular permeability, caveolin-1 and VE-cadherin, were significantly activated at 24 h in the lung tissues following CPB, which correlated with the alterations in pulmonary microvascular permeability and Src kinase. PP2 administration inhibited their activation, suggesting that they are downstream factors of Src kinase activation. The data indicated that the Src kinase pathway increased pulmonary microvascular permeability following CPB, and the activation of caveolin-1 and VE-cadherin may be involved. Inhibition of this pathway may provide a potential therapy for acute lung injury following cardiac surgery.
Collapse
Affiliation(s)
- Junwen Zhang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Zhaolei Jiang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Chunrong Bao
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Ju Mei
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Jiaquan Zhu
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| |
Collapse
|
41
|
Orhan N, Ugur Yilmaz C, Ekizoglu O, Ahishali B, Kucuk M, Arican N, Elmas I, Gürses C, Kaya M. Effects of beta-hydroxybutyrate on brain vascular permeability in rats with traumatic brain injury. Brain Res 2015; 1631:113-26. [PMID: 26656066 DOI: 10.1016/j.brainres.2015.11.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 10/29/2015] [Accepted: 11/23/2015] [Indexed: 12/16/2022]
Abstract
This study investigates the effect of beta-hydroxybutyrate (BHB) on blood-brain barrier (BBB) integrity during traumatic brain injury (TBI) in rats. Evans blue (EB) and horseradish peroxidase (HRP) were used as determinants of BBB permeability. Glutathione (GSH) and malondialdehyde (MDA) levels were estimated in the right (injury side) cerebral cortex of animals. The gene expression levels for occludin, glucose transporter (Glut)-1, aquaporin4 (AQP4) and nuclear factor-kappaB (NF-κB) were performed, and Glut-1 and NF-κB activities were analyzed. BHB treatment decreased GSH and MDA levels in intact animals and in those exposed to TBI (P<0.05). Glut-1 protein levels decreased in sham, BHB and TBI plus BHB groups (P<0.05). NF-κB protein levels increased in animals treated with BHB and/or exposed to TBI (P<0.05). The expression levels of occludin and AQP4 did not significantly change among experimental groups. Glut-1 expression levels increased in BHB treated and untreated animals exposed to TBI (P<0.05). While NF-κB expression levels increased in animals in TBI (P<0.01), a decrease was noticed in these animals upon BHB treatment (P<0.01). In animals exposed to TBI, EB extravasation was observed in the ipsilateral cortex regardless of BHB treatment. Ultrastructurally, BHB attenuated but did not prevent the presence of HRP in brain capillary endothelial cells of animals with TBI; moreover, the drug also led to the observation of the tracer when used in intact rats (P<0.01). Altogether, these results showed that BHB not only failed to provide overall protective effects on BBB in TBI but also led to BBB disruption in healthy animals.
Collapse
Affiliation(s)
- Nurcan Orhan
- Department of Neuroscience, Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Canan Ugur Yilmaz
- Department of Laboratory Animals Science, Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Department of Physiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Oguzhan Ekizoglu
- Department of Forensic Medicine, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Bulent Ahishali
- Department of Histology and Embryology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mutlu Kucuk
- Department of Laboratory Animals Science, Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Nadir Arican
- Department of Forensic Medicine, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Imdat Elmas
- Department of Forensic Medicine, Tepecik Training and Research Hospital, Izmir, Turkey
| | - Candan Gürses
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mehmet Kaya
- Department of Physiology, Koç University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
42
|
Zhang W, Xu Q, Wu J, Zhou X, Weng J, Xu J, Wang W, Huang Q, Guo X. Role of Src in Vascular Hyperpermeability Induced by Advanced Glycation End Products. Sci Rep 2015; 5:14090. [PMID: 26381822 PMCID: PMC4585381 DOI: 10.1038/srep14090] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/18/2015] [Indexed: 12/20/2022] Open
Abstract
The disruption of microvascular barrier in response to advanced glycation end products (AGEs) stimulation contributes to vasculopathy associated with diabetes mellitus. Here, to study the role of Src and its association with moesin, VE-cadherin and focal adhesion kinase (FAK) in AGE-induced vascular hyperpermeability, we verified that AGE induced phosphorylation of Src, causing increased permeability in HUVECs. Cells over-expressed Src displayed a higher permeability after AGE treatment, accompanied with more obvious F-actin rearrangement. Activation of Src with pcDNA3/flag-SrcY530F alone duplicated these effects. Inhibition of Src with siRNA, PP2 or pcDNA3/flag-SrcK298M abolished these effects. The pulmonary microvascular endothelial cells (PMVECs) isolated from receptor for AGEs (RAGE)-knockout mice decreased the phosphorylation of Src and attenuated the barrier dysfunction after AGE-treatment. In vivo study showed that the exudation of dextran from mesenteric venules was increased in AGE-treated mouse. This was attenuated in RAGE knockout or PP2-pretreated mice. Up-regulation of Src activity induced the phosphorylation of moesin, as well as activation and dissociation of VE-cadherin, while down-regulation of Src abolished these effects. FAK was also proved to interact with Src in HUVECs stimulated with AGEs. Our studies demonstrated that Src plays a critical role in AGE-induced microvascular hyperpermeability by phosphorylating moesin, VE-cadherin, and FAK respectively.
Collapse
Affiliation(s)
- Weijin Zhang
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Qiulin Xu
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, 510010, China.,Postdoctoral Workstation, Huabo Bio-pharmaceutical Research Institute, Guangzhou 510515, China
| | - Jie Wu
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyan Zhou
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Jie Weng
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Jing Xu
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Weiju Wang
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Qiaobing Huang
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Xiaohua Guo
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
43
|
Zhang J, Yang GM, Zhu Y, Peng XY, Li T, Liu LM. Role of connexin 43 in vascular hyperpermeability and relationship to Rock1-MLC20 pathway in septic rats. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1323-32. [PMID: 26342084 DOI: 10.1152/ajplung.00016.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 08/31/2015] [Indexed: 12/27/2022] Open
Abstract
Connexin (Cx)43 has been shown to participate in several cardiovascular diseases. Increased vascular permeability is a common and severe complication in sepsis or septic shock. Whether or not Cx43 takes part in the regulation of vascular permeability in severe sepsis is not known, and the underlying mechanism has not been described. With cecal ligation and puncture-induced sepsis in rats and lipopolysaccharide (LPS)-treated vascular endothelial cells (VECs) from pulmonary veins, the role of Cx43 in increased vascular permeability and its relationship to the RhoA/Rock1 pathway were studied. It was shown that vascular permeability in the lungs, kidneys, and mesentery in sepsis rats and LPS-stimulated monolayer pulmonary vein VECs was significantly increased and positively correlated with the increased expression of Cx43 and Rock1 in these organs and cultured pulmonary vein VECs. The connexin inhibitor carbenoxolone (10 mg/kg iv) and the Rock1 inhibitor Y-27632 (2 mg/kg iv) alleviated the vascular leakage of lung, mesentery, and kidney in sepsis rats. Overexpressed Cx43 increased the phosphorylation of 20-kDa myosin light chain (MLC20) and the expression of Rock1 and increased the vascular permeability and decreased the transendothelial electrical resistance of pulmonary vein VECs. Cx43 RNA interference decreased the phosphorylation of MLC20 and the expression of Rock1 and decreased LPS-stimulated hyperpermeability of cultured pulmonary vein VECs. The Rock1 inhibitor Y-27632 alleviated LPS- and overexpressed Cx43-induced hyperpermeability of monolayer pulmonary vein VECs. This report shows that Cx43 participates in the regulation of vascular permeability in sepsis and that the mechanism is related to the Rock1-MLC20 phosphorylation pathway.
Collapse
Affiliation(s)
- Jie Zhang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Guang-Ming Yang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Xiao-Yong Peng
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Tao Li
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
44
|
Wang N, Zhang D, Sun G, Zhang H, You Q, Shao M, Yue Y. Lipopolysaccharide-induced caveolin-1 phosphorylation-dependent increase in transcellular permeability precedes the increase in paracellular permeability. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:4965-77. [PMID: 26357463 PMCID: PMC4560510 DOI: 10.2147/dddt.s77646] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Lipopolysaccharide (LPS) was shown to induce an increase in caveolin-1 (Cav-1) expression in endothelial cells; however, the mechanisms regarding this response and the consequences on caveolae-mediated transcellular transport have not been completely investigated. This study aims to investigate the role of LPS-induced Cav-1 phosphorylation in pulmonary microvascular permeability in pulmonary microvascular endothelial cells (PMVECs). Methods Rat PMVECs were isolated, cultured, and identified. Endocytosis experiments were employed to stain the nuclei by DAPI, and images were obtained with a fluorescence microscope. Permeability of endothelial cultures was measured to analyze the barrier function of endothelial monolayer. Western blot assay was used to examine the expression of Cav-1, pCav-1, triton-insoluble Cav-1, and triton-soluble Cav-1 protein. Results The LPS treatment induced phosphorylation of Cav-1, but did not alter the total Cav-1 level till 60 min in both rat and human PMVECs. LPS treatment also increased the triton-insoluble Cav-1 level, which peaked 15 min after LPS treatment in both rat and human PMVECs. LPS treatment increases the intercellular cell adhesion molecule-1 expression. Src inhibitors, including PP2, PP1, Saracatinib, and Quercetin, partially inhibited LPS-induced phosphorylation of Cav-1. In addition, both PP2 and caveolae disruptor MβCD inhibited LPS-induced increase of triton-insoluble Cav-1. LPS induces permeability by activating interleukin-8 and vascular endothelial growth factor and targeting other adhesion markers, such as ZO-1 and occludin. LPS treatment also significantly increased the endocytosis of albumin, which could be blocked by PP2 or MβCD. Furthermore, LPS treatment for 15 min significantly elevated Evans Blue-labeled BSA transport in advance of a decrease in transendothelial electrical resistance of PMVEC monolayer at this time point. After LPS treatment for 30 min, transendothelial electrical resistance decreased significantly. Moreover, PP2 and MβCD blocked LPS-induced increase in Evans Blue-labeled BSA level. Conclusion Our study demonstrates that LPS-induced Cav-1 phosphorylation may lead to the increase of transcellular permeability prior to the increase of paracellular permeability in a Src-dependent manner. Thus, LPS-induced Cav-1 phosphorylation may be a therapeutic target for the treatment of inflammatory lung disease associated with elevated microvascular permeability.
Collapse
Affiliation(s)
- Nan Wang
- Department of Respiration, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China ; Department of Emergency, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Dan Zhang
- Department of Respiration, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China ; Department of Emergency, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Gengyun Sun
- Department of Respiration, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Hong Zhang
- Department of Respiration, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China ; Department of Emergency, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Qinghai You
- Department of Respiration, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Min Shao
- Department of Respiration, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Yang Yue
- Department of Respiration, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
45
|
Lin S, Nadeau PE, Mergia A. HIV inhibits endothelial reverse cholesterol transport through impacting subcellular Caveolin-1 trafficking. Retrovirology 2015; 12:62. [PMID: 26169283 PMCID: PMC4501058 DOI: 10.1186/s12977-015-0188-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/06/2015] [Indexed: 01/13/2023] Open
Abstract
Background Human immunodeficiency virus (HIV) infection leads to decreased reverse cholesterol transport (RCT) in macrophages, and Nef mediated down-regulation and redistribution of ATP-binding cassette transporter A1 (ABCA1) are identified as key factors for this effect. This may partially explain the increased risk of atherosclerosis in HIV infected individuals. Since endothelial dysfunction is key in the initial stages of atherosclerosis, we sought to determine whether RCT was affected in human aortic endothelial cells (HAECs). Results We found that apoA-I does not significantly stimulate cholesterol efflux in HAECs while cholesterol efflux to high-density lipoprotein (HDL) was dramatically reduced in HAECs co-cultured with HIV infected cells. Studies with wild type and Nef defective HIV revealed no significant differences suggesting that multiple factors are working perhaps in concert with Nef to affect cholesterol efflux to HDL from HAECs. Interestingly, treating HAECs with recombinant Nef showed similar effect in HDL mediated cholesterol efflux as observed in HAECs co-cultured with HIV infected cells. Using a detergent-free based subcellular fractionation approach, we demonstrated that exposure of HAECs to HIV infected cells or Nef alone disrupts caveolin 1 (Cav-1) subcellular trafficking upon HDL stimulation. Moreover, Nef significantly enhanced tyrosine 14 phosphorylation of Cav-1 which may have an impact on recycling of Cav-1 and caveolae. Conclusion These results suggest that HIV interferes with cholesterol efflux by HDL in HAECs through the disruption of Cav-1s’ cellular distribution and that multiple factors are involved, possibly including Nef, for the inhibition of HDL mediated cholesterol efflux and alteration of cellular distribution of Cav-1. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0188-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shanshan Lin
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, 32611, USA.
| | - Peter E Nadeau
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, 32611, USA.
| | - Ayalew Mergia
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
46
|
Sarelius IH, Glading AJ. Control of vascular permeability by adhesion molecules. Tissue Barriers 2015; 3:e985954. [PMID: 25838987 DOI: 10.4161/21688370.2014.985954] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/05/2014] [Indexed: 12/13/2022] Open
Abstract
Vascular permeability is a vital function of the circulatory system that is regulated in large part by the limited flux of solutes, water, and cells through the endothelial cell layer. One major pathway through this barrier is via the inter-endothelial junction, which is driven by the regulation of cadherin-based adhesions. The endothelium also forms attachments with surrounding proteins and cells via 2 classes of adhesion molecules, the integrins and IgCAMs. Integrins and IgCAMs propagate activation of multiple downstream signals that potentially impact cadherin adhesion. Here we discuss the known contributions of integrin and IgCAM signaling to the regulation of cadherin adhesion stability, endothelial barrier function, and vascular permeability. Emphasis is placed on known and prospective crosstalk signaling mechanisms between integrins, the IgCAMs- ICAM-1 and PECAM-1, and inter-endothelial cadherin adhesions, as potential strategic signaling nodes for multipartite regulation of cadherin adhesion.
Collapse
Key Words
- ICAM-1
- ICAM-1, intercellular adhesion molecule 1
- IgCAM, immunoglobulin superfamily cell adhesion molecule
- JAM, junctional adhesion molecule
- LPS, lipopolysaccharide
- PECAM-1
- PECAM-1, platelet endothelial cell adhesion molecule 1
- PKC, protein kinase C
- RDG, arginine-aspartic acid- glutamine
- S1P, sphingosine 1 phosphate
- SHP-2, Src homology region 2 domain-containing phosphatase
- TGF-β, transforming growth factor-β
- TNF-α, tumor necrosis factor α
- VCAM-1, vascular cell adhesion molecule 1
- VE-PTP, Receptor-type tyrosine-protein phosphatase β
- VE-cadherin
- VEGF, vascular endothelial growth factor
- adhesion
- eNOS, endothelial nitric oxide synthase
- endothelial barrier function
- fMLP, f-Met-Leu-Phe
- iNOS, inducible nitric oxide synthase
- integrins
- permeability
- transendothelial migration
Collapse
Affiliation(s)
- Ingrid H Sarelius
- University of Rochester; Department of Pharmacology and Physiology ; Rochester, NY USA
| | - Angela J Glading
- University of Rochester; Department of Pharmacology and Physiology ; Rochester, NY USA
| |
Collapse
|
47
|
Muradashvili N, Benton RL, Saatman KE, Tyagi SC, Lominadze D. Ablation of matrix metalloproteinase-9 gene decreases cerebrovascular permeability and fibrinogen deposition post traumatic brain injury in mice. Metab Brain Dis 2015; 30:411-26. [PMID: 24771110 PMCID: PMC4213324 DOI: 10.1007/s11011-014-9550-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/15/2014] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is accompanied with enhanced matrix metalloproteinase-9 (MMP-9) activity and elevated levels of plasma fibrinogen (Fg), which is a known inflammatory agent. Activation of MMP-9 and increase in blood content of Fg (i.e. hyperfibrinogenemia, HFg) both contribute to cerebrovascular disorders leading to blood brain barrier disruption. It is well-known that activation of MMP-9 contributes to vascular permeability. It has been shown that at an elevated level (i.e. HFg) Fg disrupts blood brain barrier. However, mechanisms of their actions during TBI are not known. Mild TBI was induced in wild type (WT, C57BL/6 J) and MMP-9 gene knockout (Mmp9(-/-)) homozygous, mice. Pial venular permeability to fluorescein isothiocyanate-conjugated bovine serum albumin in pericontusional area was observed 14 days after injury. Mice memory was tested with a novel object recognition test. Increased expression of Fg endothelial receptor intercellular adhesion protein-1 and formation of caveolae were associated with enhanced activity of MMP-9 causing an increase in pial venular permeability. As a result, an enhanced deposition of Fg and cellular prion protein (PrP(C)) were found in pericontusional area. These changes were attenuated in Mmp9(-/-) mice and were associated with lesser loss of short-term memory in these mice than in WT mice. Our data suggest that mild TBI-induced increased cerebrovascular permeability enhances deposition of Fg-PrP(C) and loss of memory, which is ameliorated in the absence of MMP-9 activity. Thus, targeting MMP-9 activity and blood level of Fg can be a possible therapeutic remedy to diminish vasculo-neuronal damage after TBI.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - Richard L. Benton
- Department of Anatomical Sciences and Neurobiology and Kentucky Spinal Cord Injury Research Center (KSCIRC), University of Louisville, School of Medicine, Louisville, KY
| | - Kathryn E. Saatman
- Department of Physiology and Neurosurgery and Spinal Cord & Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY, USA
| | - Suresh C. Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - David Lominadze
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
- Corresponding Author: David Lominadze, Ph. D., University of Louisville, Dept. of Physiology & Biophysics, School of Medicine, Bldg. A, Room 1115, 500 South Preston Street, Louisville, KY 40202, Phone (502) 852-4902, Fax (502) 852-6239,
| |
Collapse
|
48
|
Chignalia AZ, Vogel SM, Reynolds AB, Mehta D, Dull RO, Minshall RD, Malik AB, Liu Y. p120-catenin expressed in alveolar type II cells is essential for the regulation of lung innate immune response. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1251-63. [PMID: 25773174 DOI: 10.1016/j.ajpath.2015.01.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 12/31/2014] [Accepted: 01/22/2015] [Indexed: 10/23/2022]
Abstract
The integrity of the lung alveolar epithelial barrier is required for the gas exchange and is important for immune regulation. Alveolar epithelial barrier is composed of flat type I cells, which make up approximately 95% of the gas-exchange surface, and cuboidal type II cells, which secrete surfactants and modulate lung immunity. p120-catenin (p120; gene symbol CTNND1) is an important component of adherens junctions of epithelial cells; however, its function in lung alveolar epithelial barrier has not been addressed in genetic models. Here, we created an inducible type II cell-specific p120-knockout mouse (p120EKO). The mutant lungs showed chronic inflammation, and the alveolar epithelial barrier was leaky to (125)I-albumin tracer compared to wild type. The mutant lungs also demonstrated marked infiltration of inflammatory cells and activation of NF-κB. Intracellular adhesion molecule 1, Toll-like receptor 4, and macrophage inflammatory protein 2 were all up-regulated. p120EKO lungs showed increased expression of the surfactant proteins Sp-B, Sp-C, and Sp-D, and displayed severe inflammation after pneumonia caused by Pseudomonas aeruginosa compared with wild type. In p120-deficient type II cell monolayers, we observed reduced transepithelial resistance compared to control, consistent with formation of defective adherens junctions. Thus, although type II cells constitute only 5% of the alveolar surface area, p120 expressed in these cells plays a critical role in regulating the innate immunity of the entire lung.
Collapse
Affiliation(s)
- Andreia Z Chignalia
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Stephen M Vogel
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | | | - Dolly Mehta
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Randal O Dull
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois
| | - Richard D Minshall
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois; Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Yuru Liu
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois; University of Illinois Cancer Center, Chicago, Illinois.
| |
Collapse
|
49
|
Takano M, Kawami M, Aoki A, Yumoto R. Receptor-mediated endocytosis of macromolecules and strategy to enhance their transport in alveolar epithelial cells. Expert Opin Drug Deliv 2014; 12:813-25. [DOI: 10.1517/17425247.2015.992778] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
50
|
Curcumin ameliorates epithelial-to-mesenchymal transition of podocytes in vivo and in vitro via regulating caveolin-1. Biomed Pharmacother 2014; 68:1079-88. [DOI: 10.1016/j.biopha.2014.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/06/2014] [Indexed: 11/15/2022] Open
|