1
|
Dione MN, Zhang Q, Shang S, Lu X. Transcriptomic Analysis of Blood Collagen-Induced Arthritis Mice Exposed to 0.1 THz Reveals Inhibition of Genes and Pathways Involved in Rheumatoid Arthritis. Int J Mol Sci 2024; 25:12812. [PMID: 39684524 DOI: 10.3390/ijms252312812] [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: 10/13/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Inflammation plays an essential role in the phases of rheumatoid arthritis (RA) as the joints secrete a range of molecules that modulate the inflammatory process. While therapies based on physical properties have shown effectiveness in a range of animal experimental models, the understanding of their biological mechanisms remains unclear. The aim of this study was to investigate the immunomodulatory effects of a 0.1 terahertz (THz) wave in rheumatoid arthritis in an attempt to dissect the molecular pathways implicated. The collagen-induced rheumatoid arthritis (CIA) model joint mice were irradiated daily for 30 min over a period of 2 weeks with continuous 0.1 terahertz waves. High-throughput bulk RNA sequencing of the murine blood was performed to analyze and characterize the differences in gene expression changes between the control (Ctrl), CIA (RA), and CIA exposed to THz. Differentially expressed genes, canonical pathway analysis, gene set enrichment, and protein-protein interaction were further run on the selected DEGs. We found that terahertz exposure downregulated gene ontologies representing the "TGF-β signaling pathway", "apoptosis", "activation of T cell receptor signaling pathway", and "non-canonical NF-κB signal transduction". These observations were further confirmed by a decreased level in the expression of transcription factors Nfib and Nfatc3, and an increased level of Lsp1. In addition, the expression of Mmp8 was significantly restored. These results indicate that THz ultimately attenuates the inflammatory response of hemocytes through the T cell and NF-κB pathway, and these changes are reverberated in the blood transcriptome. In this first report of transcriptome sequencing in a model of rheumatoid arthritis exposed to terahertz waves, the downregulated DEGs were associated with anti-inflammatory effects.
Collapse
Affiliation(s)
- Mactar Ndiaga Dione
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Qi Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Sen Shang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiaoyun Lu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
2
|
Sharma P, Sethi RS. In Vivo Exposure of Deltamethrin Dysregulates the NFAT Signalling Pathway and Induces Lung Damage. J Toxicol 2024; 2024:5261994. [PMID: 39239465 PMCID: PMC11377118 DOI: 10.1155/2024/5261994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/23/2024] [Accepted: 08/10/2024] [Indexed: 09/07/2024] Open
Abstract
Deltamethrin is an insecticide used to control harmful agricultural insects that otherwise damage crops and to control vector-borne diseases. Long-term exposure to deltamethrin results in the inflammation of the lungs. The present study elucidates the molecular mechanism underlying the deltamethrin-induced lung damage. The lung samples were extracted from the Swiss albino mice following the treatment of low (2.5 mg/kg) and high (5 mg/kg) doses of deltamethrin. The mRNA expression of TCR, IL-4, and IL-13 showed upregulation, while the expression of NFAT and FOS was downregulated following a low dose of deltamethrin. Moreover, the expression of TCR was downregulated with the exposure of a high dose of deltamethrin. Furthermore, the immunohistochemistry data confirmed the pattern of protein expression for TCR, FOS, IL-4, and IL-13 following a low dose of deltamethrin exposure. However, no change was seen in the TCR, NFAT, FOS, JUN, IL-4, and IL-13 immunopositive cells of the high-dose treatment group. Also, ELISA results showed increased expression of IL-13 in the BAL fluid of animals exposed to low doses of deltamethrin. Overall, the present study showed that deltamethrin exposure induces lung damage and immune dysregulation via dysregulating the NFAT signalling pathway.
Collapse
Affiliation(s)
- Prakriti Sharma
- Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - R S Sethi
- Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| |
Collapse
|
3
|
Karpurapu M, Nie Y, Chung S, Yan J, Dougherty P, Pannu S, Wisler J, Harkless R, Parinandi N, Berdyshev E, Pei D, Christman JW. The calcineurin-NFATc pathway modulates the lipid mediators in BAL fluid extracellular vesicles, thereby regulating microvascular endothelial cell barrier function. Front Physiol 2024; 15:1378565. [PMID: 38812883 PMCID: PMC11133699 DOI: 10.3389/fphys.2024.1378565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024] Open
Abstract
Extracellular vesicles mediate intercellular communication by transporting biologically active macromolecules. Our prior studies have demonstrated that the nuclear factor of activated T cell cytoplasmic member 3 (NFATc3) is activated in mouse pulmonary macrophages in response to lipopolysaccharide (LPS). Inhibition of NFATc3 activation by a novel cell-permeable calcineurin peptide inhibitor CNI103 mitigated the development of acute lung injury (ALI) in LPS-treated mice. Although pro-inflammatory lipid mediators are known contributors to lung inflammation and injury, it remains unclear whether the calcineurin-NFATc pathway regulates extracellular vesicle (EV) lipid content and if this content contributes to ALI pathogenesis. In this study, EVs from mouse bronchoalveolar lavage fluid (BALF) were analyzed for their lipid mediators by liquid chromatography in conjunction with mass spectrometry (LC-MS/MS). Our data demonstrate that EVs from LPS-treated mice contained significantly higher levels of arachidonic acid (AA) metabolites, which were found in low levels by prior treatment with CNI103. The catalytic activity of lung tissue cytoplasmic phospholipase A2 (cPLA2) increased during ALI, correlating with an increased amount of arachidonic acid (AA) in the EVs. Furthermore, ALI is associated with increased expression of cPLA2, cyclooxygenase 2 (COX2), and lipoxygenases (5-LOX, 12-LOX, and 15-LOX) in lung tissue, and pretreatment with CNI103 inhibited the catalytic activity of cPLA2 and the expression of cPLA2, COX, and LOX transcripts. Furthermore, co-culture of mouse pulmonary microvascular endothelial cell (PMVEC) monolayer and NFAT-luciferase reporter macrophages with BALF EVs from LPS-treated mice increased the pulmonary microvascular endothelial cell (PMVEC) monolayer barrier permeability and luciferase activity in macrophages. However, EVs from CNI103-treated mice had no negative impact on PMVEC monolayer barrier integrity. In summary, BALF EVs from LPS-treated mice carry biologically active NFATc-dependent, AA-derived lipids that play a role in regulating PMVEC monolayer barrier function.
Collapse
Affiliation(s)
- Manjula Karpurapu
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Sangwoon Chung
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Jiasheng Yan
- Department of Pharmacology, Ohio State University, Columbus, OH, United States
| | - Patrick Dougherty
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - Sonal Pannu
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Jon Wisler
- Department of Surgery, Ohio State Wexner Medical Center, Columbus, OH, United States
| | - Ryan Harkless
- Department of Surgery, Ohio State Wexner Medical Center, Columbus, OH, United States
| | - Narasimham Parinandi
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Evgeny Berdyshev
- Division of Pulmonary Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - John W. Christman
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| |
Collapse
|
4
|
Halici Z, Bulut V, Cadirci E, Yayla M. Investigation of the effects of urotensin II receptors in LPS-induced inflammatory response in HUVEC cell line through calcineurin/NFATc/IL-2 pathway. Adv Med Sci 2023; 68:433-440. [PMID: 37913738 DOI: 10.1016/j.advms.2023.10.008] [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: 01/13/2023] [Revised: 07/03/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
PURPOSE The effect of urotensin II (U-II), a powerful endogenous vasoconstrictor substance, on the immune system and its mediators is very important. It was herein aimed to demonstrate the possible relationship between the calcineurin/nuclear factor of activated T-cells cytoplasmic 1/interleukin-2 (CaN/NFATc/IL-2) pathway and urotensin receptors (UTRs) in inflammatory response due to lipopolysaccharide (LPS). METHODS An LPS-induced inflammation model was used on the human umbilical vein endothelial cells (HUVEC) cell line and drugs were applied accordingly, forming the following groups: Control Group, LPS Group, Agonist Group (10-8 M U-II), Antagonist Group (10-6 M palosuran), Tacrolimus (TAC) Group (10 ng/mL FK-506), Agonist + TAC Group, and Antagonist + TAC Group. Gene expression analyses were performed using real-time polymerase chain reaction (RT-PCR). RESULTS In the analysis of the cell viability at 48 and 72 h, there was a decrease in the Agonist Group, while in the Agonist + TAC Group, the cell viability increased. In the Antagonist Group, cell viability was maintained when compared to the LPS Group, while in the TAC Group, this effect was reduced. The mRNA expression levels of UTR, CaN, NFATc, IL-2 receptor (IL-2R), IL-6 and nuclear factor kappa B (NF-κB) were higher in the LPS Group than in the Control Group, and even the UTR, CaN, NFATc, IL-2R were higher with agonist administration. This effect of the agonist was shown to be completely mitigated in the presence of the CaN inhibitor. CONCLUSION U-II and its receptors can perform key functions regarding the endothelial cell damage via the CaN/NFATc/IL-2 pathway.
Collapse
Affiliation(s)
- Zekai Halici
- Department of Pharmacology, Ataturk University, Erzurum, Turkey; Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey.
| | - Vedat Bulut
- Department of Immunology, Gazi University, Ankara, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Ataturk University, Erzurum, Turkey; Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| | - Muhammed Yayla
- Department of Pharmacology, Kafkas University, Kars, Turkey
| |
Collapse
|
5
|
Nie Y, Zhai X, Li J, Sun A, Che H, Christman JW, Chai G, Zhao P, Karpurapu M. NFATc3 Promotes Pulmonary Inflammation and Fibrosis by Regulating Production of CCL2 and CXCL2 in Macrophages. Aging Dis 2023; 14:1441-1457. [PMID: 37523510 PMCID: PMC10389814 DOI: 10.14336/ad.2022.1202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/02/2022] [Indexed: 08/02/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and highly lethal inflammatory interstitial lung disease characterized by aberrant extracellular matrix deposition. Macrophage activation by cytokines released from repetitively injured alveolar epithelial cells regulates the inflammatory response, tissue remodeling, and fibrosis throughout various phases of IPF. Our previous studies demonstrate that nuclear factor of activated T cells cytoplasmic member 3 (NFATc3) regulates a wide array of macrophage genes during acute lung injury pathogenesis. However, the role of NFATc3 in IPF pathophysiology has not been previously reported. In the current study, we demonstrate that expression of NFATc3 is elevated in lung tissues and pulmonary macrophages in mice subjected to bleomycin (BLM)-induced pulmonary fibrosis and IPF patients. Remarkably, NFATc3 deficiency (NFATc3+/-) was protective in bleomycin (BLM)-induced lung injury and fibrosis. Adoptive transfer of NFATc3+/+ macrophages to NFATc3+/- mice restored susceptibility to BLM-induced pulmonary fibrosis. Furthermore, in vitro treatment with IL-33 or conditioned medium from BLM-treated epithelial cells increased production of CCL2 and CXCL2 in macrophages from NFATc3+/+ but not NFATc3+/- mice. CXCL2 promoter-pGL3 Luciferase reporter vector showed accentuated reporter activity when co-transfected with the NFATc3 expression vector. More importantly, exogenous administration of recombinant CXCL2 into NFATc3+/- mice increased fibrotic markers and exacerbated IPF phenotype in BLM treated mice. Collectively, our data demonstrate, for the first time, that NFATc3 regulates pulmonary fibrosis by regulating CCL2 and CXCL2 gene expression in macrophages.
Collapse
Affiliation(s)
- Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China.
| | - Xiaorun Zhai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China.
| | - Jiao Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China.
| | - Aijuan Sun
- Department of Pathology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China.
| | - Huilian Che
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China.
| | - John W Christman
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA.
| | - Gaoshang Chai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China.
| | - Peng Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China.
| | - Manjula Karpurapu
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA.
| |
Collapse
|
6
|
Li Y, Long Y, Zhi X, Hao H, Wang X, Liu H, Wang L. miR-339-3p promotes AT1-AA-induced vascular inflammation by upregulating NFATc3 protein expression in vascular smooth muscle cells. Acta Biochim Biophys Sin (Shanghai) 2023; 55:295-303. [PMID: 36825443 PMCID: PMC10157516 DOI: 10.3724/abbs.2023009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Vascular inflammation induced by angiotensin II-1 receptor autoantibody (AT1-AA) is involved in the occurrence and development of various cardiovascular diseases. miR-339-3p is closely related to the degree of vasodilation of aortic aneurysm and is also involved in the occurrence and development of acute pancreatitis. However, it is still unclear whether miR-339-3p influences AT1-AA-induced vascular inflammation. In this study, the role and mechanism of miR-339-3p in AT1-AA-induced vascular inflammation are studied. RT-PCR detection shows that the miR-339-3p levels in the thoracic aorta and serum exosomes of AT1-AA-positive rats are significantly increased. The miRwalk database predicts the mRNAs that miR-339-3p can bind to their 5'UTR. Subsequently, it is found that the number of genes contained in the T cell receptor pathway is high through KEGG analysis, and NFATc3 among them can promote the secretion of various inflammatory cytokines. AT1-AA-induced upregulation of miR-339-3p expression in vascular smooth muscle cells (VSMCs) can lead to a significant increase in NFATc3 protein level and promote vascular inflammation. Inhibition of miR-339-3p with antagomir-339-3p can significantly reverse AT1-AA-induced high expressions of IL-6, IL-1β and TNF-α proteins in rat thoracic aorta and VSMCs. That is, AT1-AA can upregulate the expression of miR-339-3p in VSMCs, and the increased miR-339-3p targets the 5'UTR of NFATc3 mRNA to increase the protein level of NFATc3, thereby aggravating the occurrence of vascular inflammation. These findings provide new experimental evidence for the involvement of miRNAs in regulating vascular inflammatory diseases.
Collapse
Affiliation(s)
- Yang Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Yaolin Long
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaoyan Zhi
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Haihu Hao
- Department of Orthopedics, Shanxi Bethune Hospital & Shanxi Academy of Medical Sciences, Taiyuan 030032, China
| | - Xiaohui Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Huirong Liu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Li Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
7
|
Tyler SEB, Tyler LDK. Therapeutic roles of plants for 15 hypothesised causal bases of Alzheimer's disease. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:34. [PMID: 35996065 PMCID: PMC9395556 DOI: 10.1007/s13659-022-00354-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/15/2022] [Indexed: 05/26/2023]
Abstract
Alzheimer's disease (AD) is progressive and ultimately fatal, with current drugs failing to reverse and cure it. This study aimed to find plant species which may provide therapeutic bioactivities targeted to causal agents proposed to be driving AD. A novel toolkit methodology was employed, whereby clinical symptoms were translated into categories recognized in ethnomedicine. These categories were applied to find plant species with therapeutic effects, mined from ethnomedical surveys. Survey locations were mapped to assess how this data is at risk. Bioactivities were found of therapeutic relevance to 15 hypothesised causal bases for AD. 107 species with an ethnological report of memory improvement demonstrated therapeutic activity for all these 15 causal bases. The majority of the surveys were found to reside within biodiversity hotspots (centres of high biodiversity under threat), with loss of traditional knowledge the most common threat. Our findings suggest that the documented plants provide a large resource of AD therapeutic potential. In demonstrating bioactivities targeted to these causal bases, such plants may have the capacity to reduce or reverse AD, with promise as drug leads to target multiple AD hallmarks. However, there is a need to preserve ethnomedical knowledge, and the habitats on which this knowledge depends.
Collapse
Affiliation(s)
| | - Luke D K Tyler
- School of Natural Sciences, Bangor University, Gwynedd, UK
| |
Collapse
|
8
|
Wang WB, Li JT, Hui Y, Shi J, Wang XY, Yan SG. Combination of pseudoephedrine and emodin ameliorates LPS-induced acute lung injury by regulating macrophage M1/M2 polarization through the VIP/cAMP/PKA pathway. Chin Med 2022; 17:19. [PMID: 35123524 PMCID: PMC8818149 DOI: 10.1186/s13020-021-00562-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/25/2021] [Indexed: 12/14/2022] Open
Abstract
Background Acute lung injury (ALI) is an acute multifactorial infectious disease induced by trauma, pneumonia, shock, and sepsis. This study aimed to investigate the protective effects of pseudoephedrine and emodin combined treatment in experimental ALI, as well as the mechanisms underlying the regulation of inflammation and pulmonary edema via the VIP/cAMP/PKA pathway. Methods The wistar rats were randomly divided into fifteen groups (n = 5). Rats in each group were given intragastric administration 1 h before LPS injection. Those in the control and LPS groups were given intragastric administrations of physiological saline, rats in other groups were given intragastrically administered of differential dose therapeutic agents. The rats in the LPS and treatment groups were then injected intraperitoneally with LPS (7.5 mg/kg) to induce ALI. After being treated with pseudoephedrine and emodin for 12 h, all animals were sacrifice. Anal temperatures were taken on an hourly basis for 8 h after LPS injection. Pathological examination of lung specimen was performed by H&E staining. Cytokines (IL-1β, TNF-α, IL-6, iNOS, IL-10, Arg-1, CD86, CD206, F4/80, VIP) in lung tissue were assayed by ELISA and immunofluorescence. The expression of VIP, CAMP, AQP-1, AQP-5, p-PKA, PKA, p-IκBα, IκBα, p-p65, p65, p-P38, P38, p-ERK1/2, ERK1/2, p-JNK1/2, JNK1/2 protein in lung was determined by western blotting. Results After rats being treated with pseudoephedrine + emodin, reduced of fever symptoms. The contents of inflammatory cytokines (IL-1β, TNF-α, IL-6, iNOS) were decreased and anti-inflammatory cytokines (IL-10, Arg-1) were significantly increased in serum. Pseudoephedrine + emodin treatment effectively promoted VIP cAMP and p-PKA protein expression in lung tissues, and significantly inhibited NF-κB, MAPK phosphorylation, Pseudoephedrine + emodin treatment can inhibit M1 polarization and promoted M2 polarization via the VIP/cAMP/PKA signaling pathway. Conclusions The combination of Pseudoephedrine and emodin was effective in ameliorating LPS-induced ALI in rats by inducing VIP/cAMP/PKA signaling. Inhibiting the NF-κB, MAPK inflammatory pathway, relief of pulmonary edema suppressing macrophage M1 polarization, and promoting macrophage M2 polarization.
Collapse
|
9
|
Giblin MJ, Smith TE, Winkler G, Pendergrass HA, Kim MJ, Capozzi ME, Yang R, McCollum GW, Penn JS. Nuclear factor of activated T-cells (NFAT) regulation of IL-1β-induced retinal vascular inflammation. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166238. [PMID: 34343639 PMCID: PMC8565496 DOI: 10.1016/j.bbadis.2021.166238] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/20/2022]
Abstract
Chronic low-grade retinal inflammation is an essential contributor to the pathogenesis of diabetic retinopathy (DR). It is characterized by increased retinal cell expression and secretion of a variety of inflammatory cytokines; among these, IL-1β has the reputation of being a major driver of cytokine-induced inflammation. IL-1β and other cytokines drive inflammatory changes that cause damage to retinal cells, leading to the hallmark vascular lesions of DR; these include increased leukocyte adherence, vascular permeability, and capillary cell death. Nuclear factor of activated T-cells (NFAT) is a transcriptional regulator of inflammatory cytokines and adhesion molecules and is expressed in retinal cells. Consequently, it may influence multiple pathogenic steps early in DR. We investigated the NFAT-dependency of IL-1β-induced inflammation in human Müller cells (hMC) and human retinal microvascular endothelial cells (hRMEC). Our results show that an NFAT inhibitor, Inhibitor of NFAT-Calcineurin Association-6 (INCA-6), decreased IL-1β-induced expression of IL-1β and TNFα in hMC, while having no effect on VEGF, CCL2, or CCL5 expression. We also demonstrate that INCA-6 attenuated IL-1β-induced increases of IL-1β, TNFα, IL-6, CCL2, and CCL5 (inflammatory cytokines and chemokines), and ICAM-1 and E-selectin (leukocyte adhesion molecules) expression in hRMEC. INCA-6 similarly inhibited IL-1β-induced increases in leukocyte adhesion in both hRMEC monolayers in vitro and an acute model of retinal inflammation in vivo. Finally, INCA-6 rescued IL-1β-induced permeability in both hRMEC monolayers in vitro and an acute model of retinal inflammation in vivo. Taken together, these data demonstrate the potential of NFAT inhibition to mitigate retinal inflammation secondary to diabetes.
Collapse
Affiliation(s)
- Meredith J Giblin
- Department of Cell and Developmental Biology, Vanderbilt University, United States of America.
| | - Taylor E Smith
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, United States of America
| | - Garrett Winkler
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, United States of America
| | - Hannah A Pendergrass
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, United States of America
| | - Minjae J Kim
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, United States of America
| | - Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University, United States of America
| | - Rong Yang
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, United States of America
| | - Gary W McCollum
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, United States of America
| | - John S Penn
- Department of Cell and Developmental Biology, Vanderbilt University, United States of America; Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, United States of America
| |
Collapse
|
10
|
Bobba CM, Fei Q, Shukla V, Lee H, Patel P, Putman RK, Spitzer C, Tsai M, Wewers MD, Lee RJ, Christman JW, Ballinger MN, Ghadiali SN, Englert JA. Nanoparticle delivery of microRNA-146a regulates mechanotransduction in lung macrophages and mitigates injury during mechanical ventilation. Nat Commun 2021; 12:289. [PMID: 33436554 PMCID: PMC7804938 DOI: 10.1038/s41467-020-20449-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/03/2020] [Indexed: 12/15/2022] Open
Abstract
Mechanical ventilation generates injurious forces that exacerbate lung injury. These forces disrupt lung barrier integrity, trigger proinflammatory mediator release, and differentially regulate genes and non-coding oligonucleotides including microRNAs. In this study, we identify miR-146a as a mechanosensitive microRNA in alveolar macrophages that has therapeutic potential to mitigate lung injury during mechanical ventilation. We use humanized in-vitro systems, mouse models, and biospecimens from patients to elucidate the expression dynamics of miR-146a needed to decrease lung injury during mechanical ventilation. We find that the endogenous increase in miR-146a following injurious ventilation is not sufficient to prevent lung injury. However, when miR-146a is highly overexpressed using a nanoparticle delivery platform it is sufficient to prevent injury. These data indicate that the endogenous increase in microRNA-146a during mechanical ventilation is a compensatory response that partially limits injury and that nanoparticle delivery of miR-146a is an effective strategy for mitigating lung injury during mechanical ventilation.
Collapse
Affiliation(s)
- Christopher M Bobba
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Qinqin Fei
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH, 43210, USA
| | - Vasudha Shukla
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Hyunwook Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Pragi Patel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Rachel K Putman
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Carleen Spitzer
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - MuChun Tsai
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Mark D Wewers
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Robert J Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH, 43210, USA
| | - John W Christman
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Megan N Ballinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Samir N Ghadiali
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA.
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
| | - Joshua A Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA.
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
11
|
Dougherty PG, Karpurapu M, Koley A, Lukowski JK, Qian Z, Nirujogi TS, Rusu L, Chung S, Hummon AB, Li HW, Christman JW, Pei D. A Peptidyl Inhibitor that Blocks Calcineurin-NFAT Interaction and Prevents Acute Lung Injury. J Med Chem 2020; 63:12853-12872. [PMID: 33073986 PMCID: PMC8011862 DOI: 10.1021/acs.jmedchem.0c01236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is an inflammatory lung disease with a high morbidity and mortality rate, for which no pharmacologic treatment is currently available. Our previous studies discovered that a pivotal step in the disease process is the activation of the nuclear factor of activated T cells (NFAT) c3 in lung macrophages, suggesting that inhibitors against the upstream protein phosphatase calcineurin should be effective for prevention/treatment of ARDS. Herein, we report the development of a highly potent, cell-permeable, and metabolically stable peptidyl inhibitor, CNI103, which selectively blocks the interaction between calcineurin and NFATc3, through computational and medicinal chemistry. CNI103 specifically inhibited calcineurin signaling in vitro and in vivo and exhibited a favorable pharmacokinetic profile, broad tissue distribution following different routes of administration, and minimal toxicity. Our data indicate that CNI103 is a promising novel treatment for ARDS and other inflammatory diseases.
Collapse
Affiliation(s)
- Patrick G. Dougherty
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Ave., Columbus, OH 43210, United States
- Entrada Therapeutics, 50 Northern Avenue, Boston, MA 02210, United States
| | - Manjula Karpurapu
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio 43210, United States
| | - Amritendu Koley
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Ave., Columbus, OH 43210, United States
| | - Jessica K. Lukowski
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Ziqing Qian
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Ave., Columbus, OH 43210, United States
- Entrada Therapeutics, 50 Northern Avenue, Boston, MA 02210, United States
| | - Teja Srinivas Nirujogi
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio 43210, United States
- East Liverpool City Hospital, 425 W 5th Street, East Liverpool, Ohio 43920, United States
| | - Luiza Rusu
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio 43210, United States
| | - Sangwoon Chung
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio 43210, United States
| | - Amanda B. Hummon
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Ave., Columbus, OH 43210, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus OH, 43210, United States
| | - Hao W. Li
- Columbia Center for Translational Immunology, Columbia University, 650 W. 168 Street, New York, New York 10032, United States
| | - John W. Christman
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio 43210, United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Ave., Columbus, OH 43210, United States
| |
Collapse
|
12
|
Nie Y, Nirujogi TS, Ranjan R, Reader BF, Chung S, Ballinger MN, Englert JA, Christman JW, Karpurapu M. PolyADP-Ribosylation of NFATc3 and NF-κB Transcription Factors Modulate Macrophage Inflammatory Gene Expression in LPS-Induced Acute Lung Injury. J Innate Immun 2020; 13:83-93. [PMID: 33045713 DOI: 10.1159/000510269] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 07/16/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary macrophages play a critical role in the recognition of pathogens, initiation of host defense via inflammation, clearance of pathogens from the airways, and resolution of inflammation. Recently, we have shown a pivotal role for the nuclear factor of activated T-cell cytoplasmic member 3 (NFATc3) transcription factor in modulating pulmonary macrophage function in LPS-induced acute lung injury (ALI) pathogenesis. Although the NFATc proteins are activated primarily by calcineurin-dependent dephosphorylation, here we show that LPS induces posttranslational modification of NFATc3 by polyADP-ribose polymerase 1 (PARP-1)-mediated polyADP-ribosylation. ADP-ribosylated NFATc3 showed increased binding to iNOS and TNFα promoter DNA, thereby increasing downstream gene expression. Inhibitors of PARP-1 decreased LPS-induced NFATc3 ribosylation, target gene promoter binding, and gene expression. LPS increased NFAT luciferase reporter activity in lung macrophages and lung tissue that was inhibited by pretreatment with PARP-1 inhibitors. More importantly, pretreatment of mice with the PARP-1 inhibitor olaparib markedly decreased LPS-induced cytokines, protein extravasation in bronchoalveolar fluid, lung wet-to-dry ratios, and myeloperoxidase activity. Furthermore, PARP-1 inhibitors decreased NF-кB luciferase reporter activity and LPS-induced ALI in NF-кB reporter mice. Thus, our study demonstrates that inhibiting NFATc3 and NF-кB polyADP-ribosylation with PARP-1 inhibitors prevented LPS-induced ALI pathogenesis.
Collapse
Affiliation(s)
- Yunjuan Nie
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Teja Srinivas Nirujogi
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,East Liverpool City Hospital, East Liverpool, Ohio, USA
| | - Ravi Ranjan
- Genomics Resource Laboratory, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Brenda F Reader
- Comprehensive Transplant Center, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Sangwoon Chung
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Megan N Ballinger
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Joshua A Englert
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - John W Christman
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Manjula Karpurapu
- Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, Ohio, USA,
| |
Collapse
|
13
|
Wen L, Javed TA, Dobbs AK, Brown R, Niu M, Li L, Khalid A, Barakat MT, Xiao X, Yimlamai D, Konnikova L, Yu M, Byersdorfer CA, Husain SZ. The Protective Effects of Calcineurin on Pancreatitis in Mice Depend on the Cellular Source. Gastroenterology 2020; 159:1036-1050.e8. [PMID: 32445858 PMCID: PMC7502475 DOI: 10.1053/j.gastro.2020.05.051] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/30/2020] [Accepted: 05/14/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Calcineurin is a ubiquitously expressed central Ca2+-responsive signaling molecule that mediates acute pancreatitis, but little is known about its effects. We compared the effects of calcineurin expression by hematopoietic cells vs pancreas in mouse models of pancreatitis and pancreatitis-associated lung inflammation. METHODS We performed studies with mice with hematopoietic-specific or pancreas-specific deletion of protein phosphatase 3, regulatory subunit B, alpha isoform (PPP3R1, also called CNB1), in mice with deletion of CNB1 (Cnb1UBC△/△) and in the corresponding controls for each deletion of CNB1. Acute pancreatitis was induced in mice by administration of caerulein or high-pressure infusion of radiocontrast into biliopancreatic ducts; some mice were also given intraductal infusions of an adeno-associated virus vector that expressed nuclear factor of activated T -cells (NFAT)-luciferase into pancreas. Pancreas, bone marrow, liver, kidney, heart, and lung were collected and analyzed by histopathology, immunohistochemistry, and immunoblots; levels of cytokines were measured in serum. Mouse and human primary pancreatic acinar cells were transfected with a vector that expressed NFAT-luciferase and incubated with an agent that blocks interaction of NFAT with calcineurin; cells were analyzed by immunofluorescence. Calcineurin-mediated neutrophil chemotaxis and reactive oxygen species production were measured in neutrophils from mice. RESULTS Mice with hematopoietic-specific deletion of CNB1 developed the same level of local pancreatic inflammation as control mice after administration of caerulein or infusion of radiocontrast into biliopancreatic ducts. Cnb1UBC△/△ mice or mice with pancreas-specific deletion of CNB1 developed less severe pancreatitis and reduced pancreatic inflammation after administration of caerulein or infusion of radiocontrast into biliopancreatic ducts compared with control mice. NFAT was activated in pancreas of Swiss Webster mice given caerulein or infusions of radiocontrast into biliopancreatic ducts. Blocking the interaction between calcineurin and NFAT did not reduce pancreatic acinar cell necrosis in response to caerulein or infusions of radiocontrast. Mice with hematopoietic-specific deletion of CNB1 (but not mice with pancreas-specific deletion of CNB1) had reduced infiltration of lung tissues by neutrophils. Neutrophil chemotaxis and production of reactive oxygen species were decreased after incubation with a calcineurin inhibitor. CONCLUSIONS Hematopoietic and neutrophil expression of calcineurin promotes pancreatitis-associated lung inflammation, whereas pancreatic calcineurin promotes local pancreatic inflammation. The findings indicate that the protective effects of blocking or deleting calcineurin on pancreatitis are mediated by the source of its expression. This information should be used in the development of strategies to inhibit calcineurin for the prevention of pancreatitis and pancreatitis-associated lung inflammation.
Collapse
Affiliation(s)
- Li Wen
- Department of Gastroenterology and Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Tanveer A Javed
- Division of Pediatric Gastroenterology, University of Pittsburgh School of Medicine and the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Andrea K Dobbs
- Division of Blood and Marrow Transplantation and Cellular Therapies, University of Pittsburgh School of Medicine and the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Rebecca Brown
- Division of Blood and Marrow Transplantation and Cellular Therapies, University of Pittsburgh School of Medicine and the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Mengya Niu
- Department of Gastroenterology and Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liwen Li
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Asna Khalid
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, California
| | - Monique T Barakat
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, California; Department of Medicine, Stanford University, Palo Alto, California
| | - Xiangwei Xiao
- Division of Pediatric Surgery, University of Pittsburgh School of Medicine and the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Dean Yimlamai
- Division of Pediatric Gastroenterology, University of Pittsburgh School of Medicine and the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Liza Konnikova
- Division of Newborn Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine and the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Mang Yu
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, California
| | - Craig A Byersdorfer
- Division of Blood and Marrow Transplantation and Cellular Therapies, University of Pittsburgh School of Medicine and the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Sohail Z Husain
- Division of Pediatric Gastroenterology, Department of Pediatrics, Stanford University, Palo Alto, California.
| |
Collapse
|
14
|
Fang XZ, Ge YL, Chen ZY, Shu HQ, Yang YY, Yu Y, Zhou XJ, Chen L, Cui SN, Wang YX, Yao SL, Shang Y. NecroX-5 alleviate lipopolysaccharide-induced acute respiratory distress syndrome by inhibiting TXNIP/NLRP3 and NF-κB. Int Immunopharmacol 2020; 81:106257. [PMID: 32044659 DOI: 10.1016/j.intimp.2020.106257] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/04/2020] [Accepted: 01/24/2020] [Indexed: 02/07/2023]
Abstract
The activation of NLRP3 inflammasome and NF-κB pathway, associating with oxidativestress, have been implicated in the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). NecroX-5 has been reported to exhibit theeffectsofanti-oxidation and anti-stress in various diseases. However, the role of NecroX-5 in ALI has not been explicitly demonstrated. The aim of this study was to explore the therapeutic effects and potential mechanism action of NecroX-5 on ALI. Here, we found that NecroX-5 pretreatment dramatically diminished the levels of IL-1β, IL-18 and ROS in in RAW264.7 cells challenged with LPS and ATP. Furthermore, NecroX-5 suppressed the activation of NLRP3 inflammasome and NF-κB signalpathway. In addition, NecroX-5 also inhibited the thioredoxin-interacting protein (TXNIP) expression. In vivo, NecroX-5 reduced the LPS-induced lung histopathological injury, the number of TUNEL-positive cells, lung wet/dry (W/D) ratio, levels of total protein and inflammatory cytokines in the bronchoalveolar lavage fluid (BALF) in mice. Additionally, LPS-induced upregulation of myeloperoxidase (MPO), ROS production and malondialdehyde (MDA) were inhibited by NecroX-5 administration. Thus, our results demonstrate that NecroX-5 protects against LPS-induced ALI by inhibiting TXNIP/NLRP3 and NF-κB.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Department of Anesthesiology, Clinical Medical School of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, Jiangsu Province, PR China
| | - Ya-Li Ge
- Department of Anesthesiology, Clinical Medical School of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, Jiangsu Province, PR China
| | - Zhao-Yuan Chen
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hua-Qing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yi-Yi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong Universityof Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xiao-Jing Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Lin Chen
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shu-Nan Cui
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shang-Long Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong Universityof Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| |
Collapse
|
15
|
Qin Y, Guo X, Yu Y, Dong S, Yan Y, Bian X, Zhao C. Screening key genes and microRNAs in sepsis by RNA-sequencing. J Chin Med Assoc 2020; 83:41-47. [PMID: 31567880 DOI: 10.1097/jcma.0000000000000209] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sepsis is a life-threatening organ dysfunction, initiated by a dysregulated host response to infection. This study aimed to determine key genes and microRNAs (miRNAs) correlated with sepsis. METHODS Three patients with sepsis and three healthy individuals treated as controls were recruited in the current study. To identify differentially expressed mRNAs (DEmRNAs) and differentially expressed miRNAs (DEmiRNAs) between patients with sepsis and controls, RNA-sequencing and bioinformatics analysis were conducted. DEmiRNA-target DEmRNAs analysis and functional annotation of DEmiRNA-target DEmRNAs were performed. Dataset GSE46955, used to validate the expression of selected DEmRNAs, was downloaded from the Gene Expression Omnibus database. RESULTS Compared with septic patients, a total of 1199 DEmRNAs and 23 DEmiRNAs were identified. Based on DEmiRNA-target DEmRNAs analysis, hsa-miR-106b-5p (degree = 155), hsa-miR-128-3p (degree = 128), and hsa-miR-144-3p (degree = 79) were the top 3 DEmiRNAs that covered most DEmRNAs. The T cell receptor signaling pathway, pathways in cancer, FoxO signaling pathway, and influenza A were the significantly enriched Kyoto Encyclopedia of Genes and Genomes pathways of DEmiRNA-target DEmRNAs in sepsis. CONCLUSION We identified key genes and miRNAs related to sepsis. Our findings will provide new insights into understanding sepsis pathogenesis.
Collapse
Affiliation(s)
- Yanjun Qin
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xuan Guo
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yueqing Yu
- Department of Clinical Laboratory, Hebei General Hospital, Shijiazhuang, China
| | - Shimin Dong
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Yan
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaohua Bian
- Department of Emergency, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Caiyan Zhao
- Department of Infectious Disease, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
16
|
Li M, Fang XZ, Liu XT, Zheng YF, Xie YB, Ma XD, Xia Y, Shao DH. Inhibition of calcineurin/NFATc4 signaling attenuates ventilator‑induced lung injury. Mol Med Rep 2019; 21:607-614. [PMID: 31789412 PMCID: PMC6947850 DOI: 10.3892/mmr.2019.10851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Ventilator-induced lung injury (VILI) is a life-threatening condition caused by the inappropriate use of mechanical ventilation (MV). However, the precise molecular mechanism inducing the development of VILI remains to be elucidated. In the present study, it was revealed that the calcineurin/NFATc4 signaling pathway mediates the expression of adhesion molecules and proinflammatory cytokines essential for the development of VILI. The present results revealed that a high tidal volume ventilation (HV) caused lung inflammation and edema in the alveolar walls and the infiltration of inflammatory cells. The calcineurin activity and protein expression in the lungs were increased in animals with VILI, and NFATc4 translocated into the nucleus following calcineurin activation. Furthermore, the translocation of NFATc4 and lung injury were prevented by a calcineurin inhibitor (CsA). Thus, the present results highlighted the critical role of the calcineurin/NFATc4 signaling pathway in VILI and suggest that this pathway coincides with the release of ICAM-1, VCAM-1, TNF-α and IL-1β.
Collapse
Affiliation(s)
- Min Li
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Xiang-Zhi Fang
- Department of Anesthesiology, Clinical Medical School of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Xiao-Tian Liu
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Yong-Feng Zheng
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Yun-Bin Xie
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Xiao-Dong Ma
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Yan Xia
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Dong-Hua Shao
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| |
Collapse
|
17
|
Abstract
Approximately 75% of all disease-relevant human proteins, including those involved in intracellular protein-protein interactions (PPIs), are undruggable with the current drug modalities (i.e., small molecules and biologics). Macrocyclic peptides provide a potential solution to these undruggable targets because their larger sizes (relative to conventional small molecules) endow them the capability of binding to flat PPI interfaces with antibody-like affinity and specificity. Powerful combinatorial library technologies have been developed to routinely identify cyclic peptides as potent, specific inhibitors against proteins including PPI targets. However, with the exception of a very small set of sequences, the vast majority of cyclic peptides are impermeable to the cell membrane, preventing their application against intracellular targets. This Review examines common structural features that render most cyclic peptides membrane impermeable, as well as the unique features that allow the minority of sequences to enter the cell interior by passive diffusion, endocytosis/endosomal escape, or other mechanisms. We also present the current state of knowledge about the molecular mechanisms of cell penetration, the various strategies for designing cell-permeable, biologically active cyclic peptides against intracellular targets, and the assay methods available to quantify their cell-permeability.
Collapse
Affiliation(s)
- Patrick G. Dougherty
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, Ohio 43210, United States
| | - Ashweta Sahni
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, Ohio 43210, United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, Ohio 43210, United States
| |
Collapse
|
18
|
Transient receptor potential vanilloid 4 is a critical mediator in LPS mediated inflammation by mediating calcineurin/NFATc3 signaling. Biochem Biophys Res Commun 2019; 513:1005-1012. [PMID: 31005256 DOI: 10.1016/j.bbrc.2019.04.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/03/2019] [Indexed: 12/28/2022]
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) ion channel is thought to be an essential component of inflammatory response. However, its role and mechanism in regulating acute lung injury (ALI) and macrophages activation are not well characterized. In our study, we observe that blockade of TRPV4 using GSK2193874 or HC-067047 greatly improve the pneumonedema, the lung pathologic changes, the up-regulation of proinflammatory cytokines and the neutrophil infiltration in LPS-induced lung injury. In vitro, knockdown of TRPV4 in macrophages reduces the levels of pro-inflammatory cytokines, ROS production, Ca2+ concentration in cytoplasma and the activation of calcineurin/NFATc3 signaling. Importantly, change of extracellular Ca2+ in culture medium prevents LPS-induced NFATc3 nuclear translocation, up-regulation of proinflammatory cytokines and ROS production in macrophages. Inhibition of calcineurin with cyclosporine A, FK506 down-regulates the levels of NFATc3 nuclear translocation and proinflammatory cytokines expression. Our results demonstrate that TRPV4-dependent Ca2+ influx contributes to LPS-induced macrophage activation by calcineurin-NFATc3 pathway.
Collapse
|
19
|
Hu X, Liu S, Zhu J, Ni H. Dachengqi decoction alleviates acute lung injury and inhibits inflammatory cytokines production through TLR4/NF‐κB signaling pathway in vivo and in vitro. J Cell Biochem 2019; 120:8956-8964. [PMID: 30838705 DOI: 10.1002/jcb.27615] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/12/2018] [Accepted: 08/14/2018] [Indexed: 12/16/2022]
Affiliation(s)
- XingXing Hu
- Department of Emergency, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine Nanjing China
| | - Shang Liu
- Physical & Environmental Sciences University of Toronto Toronto Canada
| | - Jin Zhu
- Department of Emergency Medicine Nanjing University of Chinese Medicine Nanjing China
| | - HaiBin Ni
- Department of Emergency, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine Nanjing China
| |
Collapse
|
20
|
Abstract
Intracellular delivery of biological agents such as peptides, proteins, and nucleic acids generally rely on the endocytic pathway as the major uptake mechanism, resulting in their entrapment inside the endosome and lysosome. The recent discovery of cell-penetrating molecules of exceptionally high endosomal escape and cytosolic delivery efficiencies and elucidation of their mechanism of action represent major breakthroughs in this field. In this Topical Review, we provide an overview of the recent progress in understanding and enhancing the endosomal escape process and the new opportunities opened up by these recent findings.
Collapse
Affiliation(s)
- Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, Ohio 43210, USA
| | - Marina Buyanova
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, Ohio 43210, USA
| |
Collapse
|
21
|
Hough RF, Islam MN, Gusarova GA, Jin G, Das S, Bhattacharya J. Endothelial mitochondria determine rapid barrier failure in chemical lung injury. JCI Insight 2019; 4:124329. [PMID: 30728333 DOI: 10.1172/jci.insight.124329] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Acid aspiration, which can result from several etiologies, including postoperative complications, leads to direct contact of concentrated hydrochloric acid (HCl) with the alveolar epithelium. As a result, rapid endothelial activation induces alveolar inflammation, leading to life-threatening pulmonary edema. Because mechanisms underlying the rapid endothelial activation are not understood, here we determined responses in real time through optical imaging of alveoli of live mouse lungs. By alveolar micropuncture, we microinfused concentrated HCl in the alveolar lumen. As expected, acid contact with the epithelium caused rapid, but transient, apical injury. However, there was no concomitant membrane injury to the endothelium. Nevertheless, H2O2-mediated epithelial-endothelial paracrine signaling induced endothelial barrier failure, as detected by microvascular dextran leakage and lung water quantification. Remarkably, endothelial mitochondria regulated the barrier failure by activating uncoupling protein 2 (UCP2), thereby inducing transient mitochondrial depolarization that led to cofilin-induced actin depolymerization. Knockdown, or endothelium-targeted deletion of UCP2 expression, blocked these responses, including pulmonary edema. To our knowledge, these findings are the first to mechanistically implicate endothelial mitochondria in acid-induced barrier deterioration and pulmonary edema. We suggest endothelial UCP2 may be a therapeutic target for acid-induced acute lung injury.
Collapse
Affiliation(s)
- Rebecca F Hough
- Lung Biology Lab, Department of Medicine, and.,Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | | | | | | - Shonit Das
- Lung Biology Lab, Department of Medicine, and
| | | |
Collapse
|
22
|
Liao MJ, Lin H, He YW, Zou C. NFATc3 deficiency protects against high fat diet (HFD)-induced hypothalamus inflammation and apoptosis via p38 and JNK suppression. Biochem Biophys Res Commun 2018; 499:743-750. [PMID: 29596828 DOI: 10.1016/j.bbrc.2018.03.182] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 03/24/2018] [Indexed: 12/18/2022]
Abstract
Hypothalamic inflammation and apoptosis cause neural injury, playing an important role in metabolic syndrome development. Nuclear Factors of Activated T cells (NFATc3) show many physiological and pathological effects. However, the function of NFATc3 in high fat diet (HFD)-induced hypothalamus injury remains unknown. The wild type (WT) and NFATc3-knockout (KO) mice were subjected to HFD feeding for 16 weeks to examine NFATc3 function in vivo. Astrocytes isolated from WT or KO mice were cultured and exposed to fructose (Fru) in vitro. The liver damage, hypothalamus injury, pro-inflammatory markers, NF-κB (p65), Caspase-3 and mitogen-activated protein kinases (MAPKs) pathways were evaluated. NFATc3 was significantly up-regulated in hypothalamus from mice challenged with HFD, and in astrocytes incubated with Fru. Both in vivo and in vitro studies indicated that NFATc3-deletion attenuated metabolism syndrome, reduced inflammatory regulators expression, inactivated NF-κB (p65), Caspase-3 and p38/JNK signaling pathway. Of note, we identified that promoting p38 or JNK activation could rescue inflammatory response and apoptosis in NFATc3-KO astrocytes stimulated by Fru. Together, these findings revealed an important role of NFATc3 NFATc3 for HFD-induced metabolic syndrome and particularly hypothalamus injury, and understanding of the regulatory molecular mechanism might provide new and effective therapeutic strategies for prevention and treatment of hypothalamic damage associated with dietary obesity-associated neuroinflammation and apoptosis.
Collapse
Affiliation(s)
- Meng-Jun Liao
- Department of Anesthesiology, South China Hospital Affiliated to University of South China, Hengyang 421001, China
| | - Hua Lin
- Department of Anesthesia & surgery, BaoJi Municipal Central hospital, Baoji 721008, China
| | - Yun-Wu He
- Department of Pain, The Second Hospital Affiliated to University of South China, Hengyang 421001, China
| | - Cong Zou
- Department of Pain, The Second Hospital Affiliated to University of South China, Hengyang 421001, China.
| |
Collapse
|