1
|
Jin X, He M. Diagnostic biomarkers and miRNAs in prognosis of acute respiratory distress syndrome. Allergol Immunopathol (Madr) 2025; 53:194-200. [PMID: 40342127 DOI: 10.15586/aei.v53i3.1239] [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: 10/14/2024] [Accepted: 03/15/2025] [Indexed: 05/11/2025]
Abstract
Acute respiratory distress syndrome (ARDS) is a disease of the lung and/or extrapulmonary system characterized by acute, progressive breathing difficulty and refractory hypoxemia. After years of revision, the 2012 International Expert Conference developed a new diagnostic standard for ARDS, known as the Berlin definition, which provides good guidance on how to define and judge the disease in clinical practice. Despite the establishment of diagnostic standards and treatment improvements, ARDS mortality rate still remains high. The primary reason is that the pathophysiology has not been fully elucidated. In patients with ARDS, damage to the alveolar capillary membrane may occur, leading to increased vascular permeability and the occurrence of pulmonary edema. Therefore, exploring the pathogenesis of ARDS from the perspective of microvascular permeability and identification of effective targets may be key factors in the diagnosis and treatment of ARDS. This review presents the current literature regarding the role of miRNAs (micro ribonucleic acids) in early detection and prediction of ARDS outcome.
Collapse
Affiliation(s)
- Xian Jin
- Department of Critical Care Medicine, Jing'an District Central Hospital of Shanghai, Fudan University, Xikang Road, Shanghai, P.R. China;
| | - Mei He
- Department of Respiratory and Critical Care Medicine, Shanghai Tongji Hospital, Tongji University, Xincun Road, Shanghai, P.R. China
| |
Collapse
|
2
|
Liu C, Tian X, Wang Z, Mak JCW, Mao S, Liu TM, Zheng Y. Hydrogen-induced disruption of the airway mucus barrier enhances nebulized RNA delivery to reverse pulmonary fibrosis. SCIENCE ADVANCES 2025; 11:eadt2752. [PMID: 40238879 PMCID: PMC12002117 DOI: 10.1126/sciadv.adt2752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025]
Abstract
Nebulized RNA therapies are well suited for treating respiratory diseases, in particular pulmonary fibrosis (PF); however, effective delivery remains challenging. In this study, we present a highly efficient aerosol inhalation system that enables high levels of in vivo transfection efficiency in lung macrophages, yielding durable responses against PF. First, we established a nose-only aerosol inhalation device integrated with a hydrogen supplement system. This setup enables the precise administration of lipid nanoparticles (LNPs) at a controlled low dose, while simultaneously delivering the optimal concentration of therapeutic hydrogen gas. We further developed a hybrid lipid NP (HNP) by hybridizing a pH-dependent charge-inverting lipid film with apoptotic T cell membranes to enhance endosomal escape and trigger macrophage production of hepatocyte growth factor for lung repair. We demonstrated that the hydrogen flow-induced shear stresses disrupt the NP-mucus interaction, enhancing the deposition of aerosolized HNPs/TGFβ1 siRNA within fibrotic lung lesions, effectively blocking fibrogenic signaling pathways and offering a clinically viable strategy for combating PF.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xidong Tian
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Judith Choi Wo Mak
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences & Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau 999078, China
| |
Collapse
|
3
|
Khanna V, Singh K. MicroRNAs as promising drug delivery target to ameliorate chronic obstructive pulmonary disease using nano-carriers: a comprehensive review. Mol Cell Biochem 2025; 480:1431-1448. [PMID: 39254870 DOI: 10.1007/s11010-024-05110-0] [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: 06/19/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a deteriorating condition triggered by various factors, such as smoking, free radicals, and air pollution. This worsening disease is characterized by narrowing and thickening of airways, painful cough, and dyspnea. In COPD, numerous genes as well as microRNA (miRNA) play a significant role in the pathogenesis of the disease. Many in vivo and in vitro studies suggest that upregulation or suppression of certain miRNAs are effective treatment options for COPD. They have been proven to be more beneficial than the current symptomatic treatments, such as bronchodilators and corticosteroids. MiRNAs play a crucial role in immune cell development and regulate inflammatory responses in various tissues. MiRNA treatment thus allows for precision therapy with improved outcomes. Nanoparticle drug delivery systems such as polymeric nanoparticles, inorganic nanoparticles, dendrimers, polymeric micelles, and liposomes are an efficient method to ensure the biodistribution of the miRNAs to the target site. Identification of the right nanoparticle depending on the requirements and compatibility is essential for achieving maximum therapeutic effect. In this review, we offer a thorough comprehension of the pathology and genetics of COPD and the significance of miRNAs concerning various pathologies of the lung, as potential targets for treating the disease. The present review offers the latest insights into the nanoparticle drug delivery systems that can efficiently carry and deliver miRNA or antagomirs to the specific target site and hence help in effective management of COPD.
Collapse
Affiliation(s)
- Vamika Khanna
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
4
|
do Nascimento MF, Ferreira LRP, Vieira Junior JM, Deheinzelin D, Aparecida Santos Nussbaum AC, Toshihiro Sakamoto LH, Vasconcelos RO, Salomao R, Waisberg J, Azevedo LCP, Chammas R, Real JM. Circulating extracellular vesicles as potential biomarkers and mediators of acute respiratory distress syndrome in sepsis. Sci Rep 2025; 15:5512. [PMID: 39953195 PMCID: PMC11829037 DOI: 10.1038/s41598-025-89783-7] [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: 10/28/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
The early sequence of respiratory failure events after the onset of sepsis is still unknown. We hypothesize that the lung should signal through circulating extracellular vesicles (EVs) when it is affected by a systemic inflammatory response. Blood samples were obtained from septic patients with (n = 5) and without acute respiratory distress syndrome (ARDS) (n = 13) at 24 h of intensive care unit admission and 3 days later at Sírio-Libanês Hospital. Pulmonary-originated sepsis was not considered. The characteristics of the plasma-isolated EVs were compatible with exosomes. 48 miRNAs were evaluated by real-time PCR. Comparing all samples from patients with sepsis + ARDS to sepsis only, 9 miRNAs are transported in smaller amounts: miR-766 (-35.7, p = 0.002), miR-127 (-23.8, p = 0.001), miR-340 (-13.5, p = 0.006), miR-29b (-12.8, p = 0.001), miR-744 (-7.1, p = 0.05), miR-618 (-4.0, p = 0.02), miR-598 (-3.8, p = 0.035), miR-1260 (-2.5, p = 0.035); and miR-885-5p is expressed at higher levels (9.5; p = 0.028). In paired samples, the set of altered miRNAs is generally different (p < 0.05) between sepsis + ARDS (miR-1183,-1267,-1290,-17,-192,-199a-3p,-25,-485-3p,-518d,-720) or sepsis only (miR-148a,-193a-5p,-199a-3p,-222,-25,-340,744). Bioinformatic analysis showed that when sepsis is associated with ARDS, those differentially expressed miRNAs potentially target messenger RNAs from the Glycoprotein VI/GP6 signaling pathway. Circulating EV-miRNA cargo could be potential biomarkers of lung inflammation during sepsis in patients requiring mechanical ventilation.
Collapse
Affiliation(s)
- Marcelo Fernando do Nascimento
- Programa de pós-graduação em Ciências da Saúde, Instituto de Assistência Médica ao Servidor Público Estadual, São Paulo, Brasil
| | - Ludmila Rodrigues Pinto Ferreira
- RNA Systems Biology Laboratory, Departamento de morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | | | | | | | | | | | - Reinaldo Salomao
- Departamento de Imunologia, Universidade Federal de São Paulo, São Paulo, Brasil
| | - Jaques Waisberg
- Programa de pós-graduação em Ciências da Saúde, Instituto de Assistência Médica ao Servidor Público Estadual, São Paulo, Brasil
| | | | - Roger Chammas
- Instituto do Câncer do Estado de São Paulo, São Paulo, Brasil
| | - Juliana Monte Real
- Programa de pós-graduação em Ciências da Saúde, Instituto de Assistência Médica ao Servidor Público Estadual, São Paulo, Brasil.
- Instituto do Câncer do Estado de São Paulo, São Paulo, Brasil.
- Laboratório Neugen Soluções Diagnósticas, São Paulo, Brasil.
| |
Collapse
|
5
|
Clevenger AJ, Jha A, Moore E, Raghavan SA. Manipulating immune activity of macrophages: a materials and mechanics perspective. Trends Biotechnol 2025; 43:131-144. [PMID: 39155172 PMCID: PMC11717646 DOI: 10.1016/j.tibtech.2024.07.009] [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: 05/10/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024]
Abstract
Macrophage immune cells exist on a plastic spectrum of phenotypes governed by their physical and biochemical environment. Controlling macrophage function to facilitate immunological regeneration or fighting pathology has emerged as a therapeutic possibility. The rate-limiting step in translating macrophage immunomodulation therapies has been the absence of fundamental knowledge of how physics and biochemistry in the macrophage microenvironment converge to inform phenotype. In this review we explore recent trends in bioengineered model systems that integrate physical and biochemical variables applied to macrophage mechanosensing and plasticity. We focus on how tuning of mechanical forces and biomaterial composition orchestrate macrophage function in physiological and pathological contexts. Ultimately, a broader understanding of stimuli-responsiveness in macrophages leads to informed design for future modulatory therapies.
Collapse
Affiliation(s)
- Abigail J Clevenger
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Aakanksha Jha
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Erika Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
6
|
Dutta B, Mahanty M, Kesavalu L, Rahaman SO. Mechanisms underlying TRPV4-mediated regulation of miR-146a expression. Front Immunol 2024; 15:1437540. [PMID: 39403372 PMCID: PMC11471512 DOI: 10.3389/fimmu.2024.1437540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Persistent inflammation is a major contributor in the development of various inflammatory diseases like atherosclerosis. Our study investigates how transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel, interacts with microRNA-146a (miR-146a), within the context of inflammation and atherosclerosis. Micro-RNAs play a critical role in controlling gene expression, and miR-146a is notable for its anti-inflammatory actions. TRPV4 is activated by diverse soluble and mechanical stimuli, and often associated with inflammatory responses in various diseases. Here, we find that TRPV4 negatively regulates miR-146a expression in macrophages, especially following stimulation by lipopolysaccharides or alterations in matrix stiffness. We show that in atherosclerosis, a condition characterized by matrix stiffening, TRPV4 decreases miR-146a expression in aortic tissue macrophages. We find that TRPV4's impact on miR-146a is independent of activation of NFκB, Stat1, P38, and AKT, but is rather mediated through a mechanism involving histone deacetylation instead of DNA methylation at the miR-146a promoter site. Furthermore, we show that N-terminal residues 1 to 130 in TRPV4 is essential in suppression of miR-146a expression in LPS-stimulated macrophages. Altogether, this study identifies a regulatory mechanism of miR-146a expression by TRPV4 which may open new potential therapeutic strategies for managing inflammatory diseases.
Collapse
Affiliation(s)
- Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| | - Manisha Mahanty
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| | - Lakshmyya Kesavalu
- Department of Periodontology and Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, United States
| | - Shaik O. Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, United States
| |
Collapse
|
7
|
Gabela-Zuniga B, Shukla VC, Bobba C, Higuita-Castro N, Powell HM, Englert JA, Ghadiali SN. A micro-scale humanized ventilator-on-a-chip to examine the injurious effects of mechanical ventilation. LAB ON A CHIP 2024; 24:4390-4402. [PMID: 39161999 PMCID: PMC11407794 DOI: 10.1039/d4lc00143e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Patients with compromised respiratory function frequently require mechanical ventilation to survive. Unfortunately, non-uniform ventilation of injured lungs generates complex mechanical forces that lead to ventilator induced lung injury (VILI). Although investigators have developed lung-on-a-chip systems to simulate normal respiration, modeling the complex mechanics of VILI as well as the subsequent recovery phase is a challenge. Here we present a novel humanized in vitro ventilator-on-a-chip (VOC) model of the lung microenvironment that simulates the different types of injurious forces generated in the lung during mechanical ventilation. We used transepithelial/endothelial electrical impedance measurements to investigate how individual and simultaneous application of mechanical forces alters real-time changes in barrier integrity during and after injury. We find that compressive stress (i.e. barotrauma) does not significantly alter barrier integrity while over-distention (20% cyclic radial strain, volutrauma) results in decreased barrier integrity that quickly recovers upon removal of mechanical stress. Conversely, surface tension forces generated during airway reopening (atelectrauma), result in a rapid loss of barrier integrity with a delayed recovery relative to volutrauma. Simultaneous application of cyclic stretching (volutrauma) and airway reopening (atelectrauma), indicates that the surface tension forces associated with reopening fluid-occluded lung regions are the primary driver of barrier disruption. Thus, our novel VOC system can monitor the effects of different types of injurious forces on barrier disruption and recovery in real-time and can be used to interogate the biomechanical mechanisms of VILI.
Collapse
Affiliation(s)
- Basia Gabela-Zuniga
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Vasudha C Shukla
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher Bobba
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Heather M Powell
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio, USA
- Scientific Staff, Shriners Children's Ohio, Dayton, Ohio, USA
| | - Joshua A Englert
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Samir N Ghadiali
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
8
|
Lagier D, Zeng C, Kaczka DW, Zhu M, Grogg K, Gerard SE, Reinhardt JM, Ribeiro GCM, Rashid A, Winkler T, Vidal Melo MF. Mechanical ventilation guided by driving pressure optimizes local pulmonary biomechanics in an ovine model. Sci Transl Med 2024; 16:eado1097. [PMID: 39141699 DOI: 10.1126/scitranslmed.ado1097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/13/2024] [Accepted: 07/24/2024] [Indexed: 08/16/2024]
Abstract
Mechanical ventilation exposes the lung to injurious stresses and strains that can negatively affect clinical outcomes in acute respiratory distress syndrome or cause pulmonary complications after general anesthesia. Excess global lung strain, estimated as increased respiratory system driving pressure, is associated with mortality related to mechanical ventilation. The role of small-dimension biomechanical factors underlying this association and their spatial heterogeneity within the lung are currently unknown. Using four-dimensional computed tomography with a voxel resolution of 2.4 cubic millimeters and a multiresolution convolutional neural network for whole-lung image segmentation, we dynamically measured voxel-wise lung inflation and tidal parenchymal strains. Healthy or injured ovine lungs were evaluated as the mechanical ventilation positive end-expiratory pressure (PEEP) was titrated from 20 to 2 centimeters of water. The PEEP of minimal driving pressure (PEEPDP) optimized local lung biomechanics. We observed a greater rate of change in nonaerated lung mass with respect to PEEP below PEEPDP compared with PEEP values above this threshold. PEEPDP similarly characterized a breaking point in the relationships between PEEP and SD of local tidal parenchymal strain, the 95th percentile of local strains, and the magnitude of tidal overdistension. These findings advance the understanding of lung collapse, tidal overdistension, and strain heterogeneity as local triggers of ventilator-induced lung injury in large-animal lungs similar to those of humans and could inform the clinical management of mechanical ventilation to improve local lung biomechanics.
Collapse
Affiliation(s)
- David Lagier
- Experimental Interventional Imaging Laboratory (LIIE), European Center for Research in Medical Imaging (CERIMED), Aix Marseille University, Marseille 13005, France
- Department of Anesthesia and Critical Care, University Hospital La Timone, APHM, Marseille 13005, France
| | - Congli Zeng
- Department of Anesthesiology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| | - David W Kaczka
- Departments of Anesthesia and Radiology, University of Iowa, Iowa City, IA 52242, USA
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Min Zhu
- Guizhou University South Campus, Guiyang City 550025, China
| | - Kira Grogg
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Sarah E Gerard
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Joseph M Reinhardt
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Gabriel C Motta Ribeiro
- Biomedical Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-594, Brazil
| | - Azman Rashid
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tilo Winkler
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Marcos F Vidal Melo
- Department of Anesthesiology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| |
Collapse
|
9
|
Dutta B, Mahanty M, Kesavalu L, Rahaman SO. Mechanisms underlying TRPV4-mediated regulation of miR-146a expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587984. [PMID: 38617263 PMCID: PMC11014524 DOI: 10.1101/2024.04.03.587984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Persistent inflammation is a major contributor in the development of various inflammatory diseases like atherosclerosis. Our study investigates how transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel, interacts with microRNA-146a (miR-146a), within the context of inflammation and atherosclerosis. Micro-RNAs play a critical role in controlling gene expression, and miR-146a is notable for its anti-inflammatory actions. TRPV4 is activated by diverse soluble and mechanical stimuli, and often associated with inflammatory responses in various diseases. Here, we find that TRPV4 negatively regulates miR-146a expression in macrophages, especially following stimulation by lipopolysaccharides or alterations in matrix stiffness. We show that in atherosclerosis, a condition characterized by matrix stiffening, TRPV4 decreases miR-146a expression in aortic tissue macrophages. We find that TRPV4's impact on miR-146a is independent of activation of NFκB, Stat1, P38, and AKT, but is rather mediated through a mechanism involving histone deacetylation instead of DNA methylation at the miR-146a promoter site. Furthermore, we show that N-terminal residues 1 to 130 in TRPV4 is essential in suppression of miR-146a expression in LPS-stimulated macrophages. Altogether, this study identifies a regulatory mechanism of miR-146a expression by TRPV4 which may open new potential therapeutic strategies for managing inflammatory diseases.
Collapse
|
10
|
Gabela-Zuniga B, Shukla VC, Bobba C, Higuita-Castro N, Powell HM, Englert JA, Ghadiali SN. A Micro-scale Humanized Ventilator-on-a-Chip to Examine the Injurious Effects of Mechanical Ventilation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582200. [PMID: 38464068 PMCID: PMC10925162 DOI: 10.1101/2024.02.26.582200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Patients with compromised respiratory function frequently require mechanical ventilation to survive. Unfortunately, non-uniform ventilation of injured lungs generates complex mechanical forces that lead to ventilator induced lung injury (VILI). Although investigators have developed lung-on-a-chip systems to simulate normal respiration, modeling the complex mechanics of VILI as well as the subsequent recovery phase is a challenge. Here we present a novel humanized in vitro ventilator-on-a-chip (VOC) model of the lung microenvironment that simulates the different types of injurious forces generated in the lung during mechanical ventilation. We used transepithelial/endothelial electrical resistance (TEER) measurements to investigate how individual and simultaneous application of the different mechanical forces alters real-time changes in barrier integrity during and after injury. We find that compressive stress (i.e. barotrauma) does not significantly alter barrier integrity while over-distention (20% cyclic radial strain, volutrauma) results in decreased barrier integrity that quickly recovers upon removal of mechanical stress. Conversely, surface tension forces generated during airway reopening (atelectrauma), result in a rapid loss of barrier integrity with a delayed recovery relative to volutrauma. Simultaneous application of cyclic stretching (volutrauma) and airway reopening (atelectrauma), indicate that the surface tension forces associated with reopening fluid-occluded lung regions is the primary driver of barrier disruption. Thus, our novel VOC system can monitor the effects of different types of injurious forces on barrier disruption and recovery in real-time and can be used to identify the biomechanical mechanisms of VILI.
Collapse
|
11
|
Zhang S, Li S, Liu Q, Wei D, Huang L, Yin H, Yi M. Electroacupuncture alleviates ventilator-induced lung injury in mice by inhibiting the TLR4/NF-κB signaling pathway. BMC Anesthesiol 2024; 24:37. [PMID: 38263038 PMCID: PMC10804525 DOI: 10.1186/s12871-024-02408-w] [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: 09/01/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024] Open
Abstract
OBJECTIVE This study was aimed to explore the protective effect of electroacupuncture (EA) pretreatment at Zusanli point (ST36) on ventilation-induced lung injury (VILI) and its potential anti-inflammatory mechanism. METHODS High tidal volume ventilation was used to induce the VILI in mice, and EA pretreatment at ST36 was given for 7 consecutive days. The wet/dry ratio and pathological injury score of lung tissue, and total protein content of pulmonary alveolar lavage fluid (BALF) were detected after 4 h of mechanical ventilation (MV). Meanwhile, the expressions of TLR4 and NF- κB in lung tissue were evaluated by Western Blot, and the inflammatory factors in lung tissue were detected by ELISA. RESULTS After four hours of mechanical ventilation, mice with ventilator-induced lung injury showed significant increases in lung wet/dry ratio, tissue damage scores, and protein content in bronchoalveolar lavage fluid. Pro-inflammatory cytokines (IL-6, IL-1β, TNF-α) and TLR4/NF-κB expression levels in the lung were also markedly elevated (P < 0.05). Conversely, ST36 acupuncture point pre-treatment significantly reduced these parameters (P < 0.05). CONCLUSION EA pretreatment at ST36 could alleviate the inflammatory response for VILI via inhibiting TLR4/NF- κB pathway.
Collapse
Affiliation(s)
- Shuang Zhang
- Department of Anesthesiology, The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People's Hospital, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, Sichuan Province, China
| | - Shuji Li
- Department of Anesthesiology, North Sichuan Medical College, Chengdu Fifth People's Hospital, Chengdu, 611137, Sichuan Province, China
| | - Qingmei Liu
- Department of Anesthesiology, North Sichuan Medical College, Chengdu Fifth People's Hospital, Chengdu, 611137, Sichuan Province, China
| | - Daneng Wei
- Department of Anesthesiology, The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People's Hospital, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, Sichuan Province, China
| | - Liping Huang
- Department of Anesthesiology, The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People's Hospital, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, Sichuan Province, China
| | - Hong Yin
- Department of Anesthesiology, The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People's Hospital, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, Sichuan Province, China.
| | - Mingliang Yi
- Department of Anesthesiology, The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People's Hospital, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, Sichuan Province, China.
| |
Collapse
|
12
|
Wang Z, Chu C, Ding Y, Li Y, Lu C. Clinical significance of serum microRNA-146a and inflammatory factors in children with Mycoplasma pneumoniae pneumonia after azithromycin treatment. J Pediatr (Rio J) 2024; 100:108-115. [PMID: 37778397 PMCID: PMC10751685 DOI: 10.1016/j.jped.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the clinical significance of serum microRNA-146a and pro-inflammatory factors in children with Mycoplasma pneumoniae pneumonia after azithromycin treatment. microRNA-146a is known to regulate inflammatory responses, and excessive inflammation is a primary characteristic of MPP. METHODS Children with MPP received conventional symptomatic therapy along with intravenous administration of azithromycin for one week. Serum levels of microRNA-146a and pro-inflammatory factors were measured using RT-qPCR and ELISA kits, respectively. The correlation between microRNA-146a and pro-inflammatory factors was analyzed by the Pearson method. Pulmonary function indexes were assessed using a pulmonary function analyzer, and their correlation with microRNA-146a and pro-inflammatory factors after treatment was evaluated. Children with MPP were divided into effective and ineffective treatment groups, and the clinical significance of microRNA-146a and pro-inflammatory factors was evaluated using receiver operating characteristic curves and logistic multivariate regression analysis. RESULTS Serum microRNA-146a was downregulated in children with MPP but upregulated after azithromycin treatment, contrasting with the trend observed for pro-inflammatory factors. MicroRNA-146a showed a negative correlation with pro-inflammatory cytokines. Pulmonary function parameters were initially reduced in children with MPP, but increased after treatment, showing positive/inverse associations with microRNA-146a and pro-inflammatory factors. Higher microRNA-146a and lower pro-inflammatory factors predicted better efficacy of azithromycin treatment. MicroRNA-146a, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-8 (IL-8), and forced expiratory volume in the first second/forced vital capacity (FEV1/FVC) were identified as independent factors influencing treatment efficacy. CONCLUSION Azithromycin treatment in children with MPP upregulates microRNA-146a, downregulates pro-inflammatory factors, and effectively improves pulmonary function.
Collapse
Affiliation(s)
- Zhe Wang
- Children's Hospital of Soochow University, Department of Infectious Disease, Suzhou, Jiangsu, China
| | - Chu Chu
- Children's Hospital of Soochow University, Department of Infectious Disease, Suzhou, Jiangsu, China
| | - Ying Ding
- Children's Hospital of Soochow University, Department of Infectious Disease, Suzhou, Jiangsu, China
| | - Yuqin Li
- Children's Hospital of Soochow University, Department of Infectious Disease, Suzhou, Jiangsu, China
| | - Chunyu Lu
- Children's Hospital of Soochow University, Department of Infectious Disease, Suzhou, Jiangsu, China.
| |
Collapse
|
13
|
Ali Zaidi SS, Fatima F, Ali Zaidi SA, Zhou D, Deng W, Liu S. Engineering siRNA therapeutics: challenges and strategies. J Nanobiotechnology 2023; 21:381. [PMID: 37848888 PMCID: PMC10583313 DOI: 10.1186/s12951-023-02147-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023] Open
Abstract
Small interfering RNA (siRNA) is a potential method of gene silencing to target specific genes. Although the U.S. Food and Drug Administration (FDA) has approved multiple siRNA-based therapeutics, many biological barriers limit their use for treating diseases. Such limitations include challenges concerning systemic or local administration, short half-life, rapid clearance rates, nonspecific binding, cell membrane penetration inability, ineffective endosomal escape, pH sensitivity, endonuclease degradation, immunological responses, and intracellular trafficking. To overcome these barriers, various strategies have been developed to stabilize siRNA, ensuring their delivery to the target site. Chemical modifications implemented with nucleotides or the phosphate backbone can reduce off-target binding and immune stimulation. Encapsulation or formulation can protect siRNA from endonuclease degradation and enhance cellular uptake while promoting endosomal escape. Additionally, various techniques such as viral vectors, aptamers, cell-penetrating peptides, liposomes, and polymers have been developed for delivering siRNA, greatly improving their bioavailability and therapeutic potential.
Collapse
Affiliation(s)
- Syed Saqib Ali Zaidi
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Faria Fatima
- College of Medical Technology, Ziauddin University, Karachi, 74700, Pakistan
| | - Syed Aqib Ali Zaidi
- Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China
| | - Dezhong Zhou
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Wuquan Deng
- Department of Endocrinology and Metabolism, Chongqing Diabetic Foot Medical Research Center, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China.
| | - Shuai Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
14
|
Fei Q, Shalosky EM, Barnes R, Shukla VC, Xu S, Ballinger MN, Farkas L, Lee RJ, Ghadiali SN, Englert JA. Macrophage-Targeted Lipid Nanoparticle Delivery of microRNA-146a to Mitigate Hemorrhagic Shock-Induced Acute Respiratory Distress Syndrome. ACS NANO 2023; 17:16539-16552. [PMID: 37595605 PMCID: PMC10754353 DOI: 10.1021/acsnano.3c01814] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2023]
Abstract
The pro-inflammatory response of alveolar macrophages to injurious physical forces during mechanical ventilation is regulated by the anti-inflammatory microRNA, miR-146a. Increasing miR-146a expression to supraphysiologic levels using untargeted lipid nanoparticles reduces ventilator-induced lung injury but requires a high initial dose of miR-146a making it less clinically applicable. In this study, we developed mannosylated lipid nanoparticles that can effectively mitigate lung injury at the initiation of mechanical ventilation with lower doses of miR-146a. We used a physiologically relevant humanized in vitro coculture system to evaluate the cell-specific targeting efficiency of the mannosylated lipid nanoparticle. We discovered that mannosylated lipid nanoparticles preferentially deliver miR-146a to alveolar macrophages and reduce force-induced inflammation in vitro. Our in vivo study using a clinically relevant mouse model of hemorrhagic shock-induced acute respiratory distress syndrome demonstrated that delivery of a low dose of miR-146a (0.1 nmol) using mannosylated lipid nanoparticles dramatically increases miR-146a levels in mouse alveolar macrophages and decreases lung inflammation. These data suggest that mannosylated lipid nanoparticles may have the therapeutic potential to mitigate lung injury during mechanical ventilation.
Collapse
Affiliation(s)
- Qinqin Fei
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, USA
- 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
| | - Emily M. Shalosky
- 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
| | - Ryelie Barnes
- 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
| | - Vasudha C. Shukla
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH 43210, USA
| | - Siying Xu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 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
| | - Laszlo Farkas
- 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
| | - Samir N. Ghadiali
- 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 Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH 43210, USA
- 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
| |
Collapse
|
15
|
Han H, Xing L, Chen BT, Liu Y, Zhou TJ, Wang Y, Zhang LF, Li L, Cho CS, Jiang HL. Progress on the pathological tissue microenvironment barrier-modulated nanomedicine. Adv Drug Deliv Rev 2023; 200:115051. [PMID: 37549848 DOI: 10.1016/j.addr.2023.115051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Imbalance in the tissue microenvironment is the main obstacle to drug delivery and distribution in the human body. Before penetrating the pathological tissue microenvironment to the target site, therapeutic agents are usually accompanied by three consumption steps: the first step is tissue physical barriers for prevention of their penetration, the second step is inactivation of them by biological molecules, and the third step is a cytoprotective mechanism for preventing them from functioning on specific subcellular organelles. However, recent studies in drug-hindering mainly focus on normal physiological rather than pathological microenvironment, and the repair of damaged physiological barriers is also rarely discussed. Actually, both the modulation of pathological barriers and the repair of damaged physiological barriers are essential in the disease treatment and the homeostasis maintenance. In this review, we present an overview describing the latest advances in the generality of these pathological barriers and barrier-modulated nanomedicine. Overall, this review holds considerable significance for guiding the design of nanomedicine to increase drug efficacy in the future.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
16
|
Almeida AF, Miranda MS, Vinhas A, Rodrigues MT, Gomes ME. Contactless Resolution of Inflammatory Signals in Tailored Macrophage-Based Cell Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37527508 DOI: 10.1021/acsami.2c22505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
In recent years, nanotechnology-based microRNA (miR) therapeutic platforms have shown great promise for immunotherapy and tissue regeneration, despite the unmet challenge of achieving efficient and safe delivery of miRs. The transport of miRs offers precision and regulatory value for a myriad of biological processes and pathways, including the control of macrophage (Mφ) functions and, consequently, the inflammatory cascades Mφ are involved in. Thus, enforcement of Mφ can boost the regenerative process and provide new solutions for diverse chronic pathologies. In this study, we sought to develop a magnetically guided transporter to deliver an miR-155 antagonist to M1-primed Mφ. Furthermore, we determined its modulatory effect in reprogramming Mφ from inflammatory to pro-regenerative phenotypes, with the aim of tissue healing and regenerative medicine approaches. This strategy combines contactless and high-precision control of Mφ, anticipating new functional miR carriers for targeted strategies controlled by extracorporeal action. The magnetoplexes SPION@PEI-miR were efficiently delivered into Mφ without compromising cell viability and successfully induced miR-mediated gene silencing by enhancing the expression of anti-inflammatory markers (IL4 and IL10) and the production of M2φ-related markers (CD206 and IL4). Given its multimodal features, SPION@PEI-miR represents a simple, safe, and nonviral theranostic platform that enables imaging, tracking, and miR delivery with modulatory effects on immune cells.
Collapse
Affiliation(s)
- Ana F Almeida
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4710-057, Portugal
| | - Margarida S Miranda
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4710-057, Portugal
| | - Adriana Vinhas
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4710-057, Portugal
| | - Márcia T Rodrigues
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4710-057, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4710-057, Portugal
| |
Collapse
|
17
|
Ghafouri-Fard S, Shoorei H, Noferesti L, Hussen BM, Moghadam MHB, Taheri M, Rashnoo F. Nanoparticle-mediated delivery of microRNAs-based therapies for treatment of disorders. Pathol Res Pract 2023; 248:154667. [PMID: 37422972 DOI: 10.1016/j.prp.2023.154667] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/01/2023] [Accepted: 07/02/2023] [Indexed: 07/11/2023]
Abstract
miRNAs represent appropriate candidates for treatment of several disorders. However, safe and efficient delivery of these small-sized transcripts has been challenging. Nanoparticle-based delivery of miRNAs has been used for treatment of a variety of disorders, particularly cancers as well as ischemic stroke and pulmonary fibrosis. The wide range application of this type of therapy is based on the important roles of miRNAs in the regulation of cell behavior in physiological and pathological conditions. Besides, the ability of miRNAs to inhibit or increase expression of several genes gives them the superiority over mRNA or siRNA-based therapies. Preparation of nanoparticles for miRNA delivery is mainly achieved through using protocols originally developed for drugs or other types of biomolecules. In brief, nanoparticle-based delivery of miRNAs is regarded as a solution for overcoming all challenges in the therapeutic application of miRNAs. Herein, we provide an overview of studies which used nanoparticles as delivery systems for facilitation of miRNAs entry into target cells for the therapeutic purposes. However, our knowledge about miRNA-loaded nanoparticles is limited, and it is expected that numerous therapeutic possibilities will be revealed for miRNA-loaded nanoparticles in future.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran; Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Noferesti
- Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | | | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fariborz Rashnoo
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Delivery of anti-microRNA-21 by lung-targeted liposomes for pulmonary fibrosis treatment. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:36-47. [PMID: 36919116 PMCID: PMC9972768 DOI: 10.1016/j.omtn.2023.02.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disorder with a low survival rate. Pulmonary fibrosis is one of the complications of COVID-19 and has a high prevalence in COVID-19 patients. Currently, no effective therapies other than lung transplantation are available to cure IPF and post-COVID-19 pulmonary fibrosis. MicroRNAs are small non-coding RNAs that mediate the development and progression of pulmonary fibrosis, thus making them potent drug candidates for this serious disease. MicroRNA-21 (miR-21) promotes not only the differentiation of fibroblasts to myofibroblasts but also epithelial-mesenchymal transition, both of which have been proposed as fundamental processes in pulmonary fibrosis development. Delivery of anti-miR-21 to block the miR-21-associated fibrogenic pathways represents a promising therapy for pulmonary fibrosis. However, microRNA treatment is challenged by quick degradation of RNA in blood, poor cellular uptake, and off-target effects. To overcome these challenges, we developed a lung-targeted, cationic liposome formulation to encapsulate anti-miR-21, enhance its delivery efficiency, and improve the therapeutic efficacy. We optimized the liposome formulation and demonstrated the anti-fibrotic effects using both in vitro and in vivo lung fibrosis models. Our results showed that anti-miR-21 delivered by cationic liposomes suppressed myofibroblast differentiation, reduced the synthesis of extracellular matrix, and inhibited fibrosis progression.
Collapse
|
19
|
Wallbank AM, Vaughn AE, Niemiec S, Bilodeaux J, Lehmann T, Knudsen L, Kolanthai E, Seal S, Zgheib C, Nozik E, Liechty KW, Smith BJ. CNP-miR146a improves outcomes in a two-hit acute- and ventilator-induced lung injury model. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102679. [PMID: 37116556 PMCID: PMC10129905 DOI: 10.1016/j.nano.2023.102679] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/21/2023] [Accepted: 04/09/2023] [Indexed: 04/30/2023]
Abstract
Acute respiratory distress syndrome (ARDS) has high mortality (~40 %) and requires the lifesaving intervention of mechanical ventilation. A variety of systemic inflammatory insults can progress to ARDS, and the inflamed and injured lung is susceptible to ventilator-induced lung injury (VILI). Strategies to mitigate the inflammatory response while restoring pulmonary function are limited, thus we sought to determine if treatment with CNP-miR146a, a conjugate of novel free radical scavenging cerium oxide nanoparticles (CNP) to the anti-inflammatory microRNA (miR)-146a, would protect murine lungs from acute lung injury (ALI) induced with intratracheal endotoxin and subsequent VILI. Lung injury severity and treatment efficacy were evaluated via lung mechanical function, relative gene expression of inflammatory biomarkers, and lung morphometry (stereology). CNP-miR146a reduced the severity of ALI and slowed the progression of VILI, evidenced by improvements in inflammatory biomarkers, atelectasis, gas volumes in the parenchymal airspaces, and the stiffness of the pulmonary system.
Collapse
Affiliation(s)
- Alison M Wallbank
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
| | - Alyssa E Vaughn
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Steve Niemiec
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Jill Bilodeaux
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
| | - Tanner Lehmann
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Lars Knudsen
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research, Germany
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine and Children's Hospital Colorado, Aurora, CO, USA; Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine and Banner Children's at Diamond Children's Medical Center, Tucson, AZ, USA
| | - Eva Nozik
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatric Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Aurora, CO, USA
| | - Kenneth W Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine and Children's Hospital Colorado, Aurora, CO, USA; Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine and Banner Children's at Diamond Children's Medical Center, Tucson, AZ, USA
| | - Bradford J Smith
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA; Department of Pediatric Pulmonary and Sleep Medicine, School of Medicine, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
20
|
Lian J, Zhu X, Du J, Huang B, Zhao F, Ma C, Guo R, Zhang Y, Ji L, Yahaya BH, Lin J. Extracellular vesicle-transmitted miR-671-5p alleviates lung inflammation and injury by regulating the AAK1/NF-κB axis. Mol Ther 2023; 31:1365-1382. [PMID: 36733250 PMCID: PMC10188640 DOI: 10.1016/j.ymthe.2023.01.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/08/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem cells regulate remote intercellular signaling communication via their secreted extracellular vesicles. Here, we report that menstrual blood-derived stem cells alleviate acute lung inflammation and injury via their extracellular vesicle-transmitted miR-671-5p. Disruption of this abundantly expressed miR-671-5p dramatically reduced the ameliorative effect of extracellular vesicles released by menstrual blood-derived stem cells on lipopolysaccharide (LPS)-induced pulmonary inflammatory injury. Mechanistically, miR-671-5p directly targets the kinase AAK1 for post-transcriptional degradation. AAK1 is found to positively regulate the activation of nuclear factor κB (NF-κB) signaling by controlling the stability of the inhibitory protein IκBα. This study identifies a potential molecular basis of how extracellular vesicles derived from mesenchymal stem cells improve pulmonary inflammatory injury and highlights the functional importance of the miR-671-5p/AAK1 axis in the progression of pulmonary inflammatory diseases. More importantly, this study provides a promising cell-based approach for the treatment of pulmonary inflammatory disorders through an extracellular vesicle-dependent pathway.
Collapse
Affiliation(s)
- Jie Lian
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China; Lung Stem Cells and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, SAINS@Bertam, 13200 Kepala Batas, Penang, Malaysia; Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Xinxing Zhu
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China.
| | - Jiang Du
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Beijia Huang
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Fengting Zhao
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Chunya Ma
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Rui Guo
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Yangxia Zhang
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Longkai Ji
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Badrul Hisham Yahaya
- Lung Stem Cells and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, SAINS@Bertam, 13200 Kepala Batas, Penang, Malaysia.
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China; Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
21
|
Fei Q, Bentley I, Ghadiali SN, Englert JA. Pulmonary drug delivery for acute respiratory distress syndrome. Pulm Pharmacol Ther 2023; 79:102196. [PMID: 36682407 PMCID: PMC9851918 DOI: 10.1016/j.pupt.2023.102196] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
The acute respiratory distress syndrome (ARDS) is a life-threatening condition that causes respiratory failure. Despite numerous clinical trials, there are no molecularly targeted pharmacologic therapies to prevent or treat ARDS. Drug delivery during ARDS is challenging due to the heterogenous nature of lung injury and occlusion of lung units by edema fluid and inflammation. Pulmonary drug delivery during ARDS offers several potential advantages including limiting the off-target and off-organ effects and directly targeting the damaged and inflamed lung regions. In this review we summarize recent ARDS clinical trials using both systemic and pulmonary drug delivery. We then discuss the advantages of pulmonary drug delivery and potential challenges to its implementation. Finally, we discuss the use of nanoparticle drug delivery and surfactant-based drug carriers as potential strategies for delivering therapeutics to the injured lung in ARDS.
Collapse
Affiliation(s)
- Qinqin Fei
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH, 43210, USA; 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, 140West 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
| | - Ian Bentley
- 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
- Department of Biomedical Engineering, The Ohio State University, 140West 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; The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
22
|
Han H, Xing J, Chen W, Jia J, Li Q. Fluorinated polyamidoamine dendrimer-mediated miR-23b delivery for the treatment of experimental rheumatoid arthritis in rats. Nat Commun 2023; 14:944. [PMID: 36805456 PMCID: PMC9941585 DOI: 10.1038/s41467-023-36625-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
In rheumatoid arthritis (RA), insufficient apoptosis of macrophages and excessive generation of pro-inflammatory cytokines are intimately connected, accelerating the development of disease. Here, a fluorinated polyamidoamine dendrimer (FP) is used to deliver miR-23b to reduce inflammation by triggering the apoptosis of as well as inhibiting the inflammatory response in macrophages. Following the intravenous injection of FP/miR-23b nanoparticles in experimental RA models, the nanoparticles show therapeutic efficacy with inhibition of inflammatory response, reduced bone and cartilage erosion, suppression of synoviocyte infiltration and the recovery of mobility. Moreover, the nanoparticles accumulate in the inflamed joint and are non-specifically captured by synoviocytes, leading to the restoration of miR-23b expression in the synovium. The miR-23b nanoparticles target Tab2, Tab3 and Ikka to regulate the activation of NF-κB pathway in the hyperplastic synovium, thereby promoting anti-inflammatory and anti-proliferative responses. Additionally, the intravenous administration of FP/miR-23b nanoparticles do not induce obvious systemic toxicity. Overall, our work demonstrates that the combination of apoptosis induction and inflammatory inhibition could be a promising approach in the treatment of RA and possibly other autoimmune diseases.
Collapse
Affiliation(s)
- Haobo Han
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China
| | - Jiakai Xing
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China
| | - Wenqi Chen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China
| | - Jiaxin Jia
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 130012, Changchun, China.
| |
Collapse
|
23
|
Fei Q, Shalosky EM, Barnes R, Shukla VC, Ballinger MN, Farkas L, Lee RJ, Ghadiali SN, Englert JA. Macrophage-targeted lipid nanoparticle delivery of microRNA-146a to mitigate hemorrhagic shock-induced acute respiratory distress syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.17.529007. [PMID: 36824913 PMCID: PMC9949132 DOI: 10.1101/2023.02.17.529007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The pro-inflammatory response of alveolar macrophages to injurious physical forces during mechanical ventilation is regulated by the anti-inflammatory microRNA, miR-146a. Increasing miR-146a expression to supraphysiologic levels using untargeted lipid nanoparticles reduces ventilator-induced lung injury, but requires a high initial dose of miR-146a making it less clinically applicable. In this study, we developed mannosylated lipid nanoparticles that can effectively mitigate lung injury at the initiation of mechanical ventilation with lower doses of miR-146a. We used a physiologically relevant humanized in vitro co-culture system to evaluate the cell-specific targeting efficiency of the mannosylated lipid nanoparticle. We discovered that mannosylated lipid nanoparticles preferentially deliver miR-146a to alveolar macrophages and reduce force-induced inflammation in vitro . Our in vivo study using a clinically relevant mouse model of hemorrhagic shock-induced acute respiratory distress syndrome demonstrated that delivery of a low dose miR-146a (0.1 nmol) using mannosylated lipid nanoparticles dramatically increases miR-146a in mouse alveolar macrophages and decreases lung inflammation. These data suggest that mannosylated lipid nanoparticles may have therapeutic potential to mitigate lung injury during mechanical ventilation.
Collapse
|
24
|
Li J, Chen L, Sun H, Zhan M, Laurent R, Mignani S, Majoral JP, Shen M, Shi X. Cationic phosphorus dendron nanomicelles deliver microRNA mimics and microRNA inhibitors for enhanced anti-inflammatory therapy of acute lung injury. Biomater Sci 2023; 11:1530-1539. [PMID: 36607143 DOI: 10.1039/d2bm01807a] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of efficient nanomedicines to repress the repolarization of M1 phenotype macrophages and therefore inhibit pro-inflammatory cytokine overexpression for anti-inflammatory therapy is still a challenging task. We report here an original gene delivery nanoplatform based on pyrrolidinium-modified amphiphilic generation 1 phosphorus dendron (C12G1) nanomicelles with a rigid phosphorous dendron structure. The nanomicelles display higher gene delivery efficiency than the counterpart materials of pyrrolidinium-modified G1 phosphorus dendrimers, and meanwhile exhibit excellent cytocompatibility. The C12G1 nanomicelles can be employed to co-deliver the miRNA-146a mimic (miR-146a mimic) and miRNA-429 inhibitor (miR-429i) to inhibit the Toll-like receptor-4 signaling pathway and p38 mitogen-activated protein kinase signaling pathway, respectively, thus causing repression of M1 phenotype alveolar macrophage polarization. The developed C12G1/miR-mixture polyplexes enable efficient therapy of lipopolysaccharide-activated alveolar macrophages in vitro and an acute lung injury mouse model in vivo. The generated cationic phosphorus dendron nanomicelles may hold promising potential for anti-inflammatory gene therapy of other inflammatory diseases.
Collapse
Affiliation(s)
- Jin Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China. .,Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, BP 44099, 31077 Toulouse CEDEX 4, France
| | - Liang Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China. .,Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, BP 44099, 31077 Toulouse CEDEX 4, France
| | - Huxiao Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Regis Laurent
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, BP 44099, 31077 Toulouse CEDEX 4, France.,Université de Toulouse, UPS, INPT, 31077 Toulouse CEDEX 4, France
| | - Serge Mignani
- Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, 45, rue des Saints Pères, 75006 Paris, France.,CQM-Centro de Quimica da Madeira, Universidade da Madeira, 9020-105 Funchal, Portugal
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, BP 44099, 31077 Toulouse CEDEX 4, France.,Université de Toulouse, UPS, INPT, 31077 Toulouse CEDEX 4, France
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China. .,CQM-Centro de Quimica da Madeira, Universidade da Madeira, 9020-105 Funchal, Portugal
| |
Collapse
|
25
|
Shirvani H, Jafari H, Moravveji SS, Abbasi Faranghizadeh F, Talebi M, Ghanavi J, Esfandi F, Najafi S, Nasiri Moghadam M, Farnia P, Aghaei Zarch SM. Non-coding RNA in SARS-CoV-2: Progress toward therapeutic significance. Int J Biol Macromol 2022; 222:1538-1550. [PMID: 36152703 PMCID: PMC9492401 DOI: 10.1016/j.ijbiomac.2022.09.105] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 09/09/2022] [Accepted: 09/11/2022] [Indexed: 12/18/2022]
Abstract
The recently developed pathogenic virus, SARS-CoV-2, was found in the Hubei Province, China. Giving rise to a broad spectrum of symptoms, SARS-CoV-2 rapidly spread across the globe, causing multi-systemic and dangerous complications, with death in extreme cases. Thereby, the number of research cases increases every day on preventing infection and treating its resulting damage. Accumulating evidence suggests noncoding RNAs (ncRNAs) are necessary for modifying virus infection and antiviral immune reaction, along with biological processes regulating SARS-CoV-2 and subsequent disease states. Therefore, understanding these mechanisms might provide a further understanding of the pathogenesis and feasible therapy alternatives against SARS-CoV2. Consequently, the molecular biology of SARS-CoV-2, ncRNA's role in its infection, and various RNA therapy tactics against the virus have been presented in this review section.
Collapse
Affiliation(s)
- Hanieh Shirvani
- Biocenter Oulu and Faculity of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | | | | | | | - Mehrdad Talebi
- Department of Medical Genetics, Faculity of Medicine, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Jalaledin Ghanavi
- Mycobacteriology Research Centre, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farbod Esfandi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Poopak Farnia
- Mycobacteriology Research Centre, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
26
|
Johnson C, Exell J, Lin Y, Aguilar J, Welsher KD. Capturing the start point of the virus-cell interaction with high-speed 3D single-virus tracking. Nat Methods 2022; 19:1642-1652. [PMID: 36357694 PMCID: PMC10154077 DOI: 10.1038/s41592-022-01672-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 09/30/2022] [Indexed: 11/12/2022]
Abstract
The early stages of the virus-cell interaction have long evaded observation by existing microscopy methods due to the rapid diffusion of virions in the extracellular space and the large three-dimensional cellular structures involved. Here we present an active-feedback single-particle tracking method with simultaneous volumetric imaging of the live cell environment called 3D-TrIm to address this knowledge gap. 3D-TrIm captures the extracellular phase of the infectious cycle in what we believe is unprecedented detail. We report what are, to our knowledge, previously unobserved phenomena in the early stages of the virus-cell interaction, including skimming contact events at the millisecond timescale, orders of magnitude change in diffusion coefficient upon binding and cylindrical and linear diffusion modes along cellular protrusions. Finally, we demonstrate how this method can move single-particle tracking from simple monolayer culture toward more tissue-like conditions by tracking single virions in tightly packed epithelial cells. This multiresolution method presents opportunities for capturing fast, three-dimensional processes in biological systems.
Collapse
Affiliation(s)
| | - Jack Exell
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Yuxin Lin
- Department of Chemistry, Duke University, Durham, NC, USA
| | | | | |
Collapse
|
27
|
Aegerter H, Lambrecht BN, Jakubzick CV. Biology of lung macrophages in health and disease. Immunity 2022; 55:1564-1580. [PMID: 36103853 DOI: 10.1016/j.immuni.2022.08.010] [Citation(s) in RCA: 272] [Impact Index Per Article: 90.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 12/14/2022]
Abstract
Tissue-resident alveolar and interstitial macrophages and recruited macrophages are critical players in innate immunity and maintenance of lung homeostasis. Until recently, assessing the differential functional contributions of tissue-resident versus recruited macrophages has been challenging because they share overlapping cell surface markers, making it difficult to separate them using conventional methods. This review describes how scRNA-seq and spatial transcriptomics can separate these subpopulations and help unravel the complexity of macrophage biology in homeostasis and disease. First, we provide a guide to identifying and distinguishing lung macrophages from other mononuclear phagocytes in humans and mice. Second, we outline emerging concepts related to the development and function of the various lung macrophages in the alveolar, perivascular, and interstitial niches. Finally, we describe how different tissue states profoundly alter their functions, including acute and chronic lung disease, cancer, and aging.
Collapse
Affiliation(s)
- Helena Aegerter
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, ErasmusMC, Rotterdam, the Netherlands
| | - Claudia V Jakubzick
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA.
| |
Collapse
|
28
|
Huang H, Zhu J, Gu L, Hu J, Feng X, Huang W, Wang S, Yang Y, Cui P, Lin SH, Suen A, Shimada BK, Williams B, Kane MA, Ke Y, Zhang CO, Birukova AA, Birukov KG, Chao W, Zou L. TLR7 Mediates Acute Respiratory Distress Syndrome in Sepsis by Sensing Extracellular miR-146a. Am J Respir Cell Mol Biol 2022; 67:375-388. [PMID: 35679261 PMCID: PMC9447138 DOI: 10.1165/rcmb.2021-0551oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
TLR7 (Toll-like receptor 7), the sensor for single-stranded RNA, contributes to systemic inflammation and mortality in murine polymicrobial sepsis. Recent studies show that extracellular miR-146a-5p serves as a TLR7 ligand and plays an important role in regulating host innate immunity. However, the role of miR-146a-5p and TLR7 signaling in pulmonary inflammation, endothelial activation, and sepsis-associated acute respiratory distress syndrome remains unclear. Here, we show that intratracheal administration of exogenous miR-146a-5p in mice evokes lung inflammation, activates endothelium, and increases endothelial permeability via TLR7-dependent mechanisms. TLR7 deficiency attenuates pulmonary barrier dysfunction and reduces lung inflammatory response in a murine sepsis model. Moreover, the impact of miR-146a-5p-TLR7 signaling on endothelial activation appears to be a secondary effect because TLR7 is undetectable in the human pulmonary artery and microvascular endothelial cells (ECs), which show no response to direct miR-146a-5p treatment in vitro. Both conditioned media of miR-146a-5p-treated macrophages (Mϕ) and septic sera of wild-type mice induce a marked EC barrier disruption in vitro, whereas Mϕ conditioned media or septic sera of TLR7-/- mice do not exhibit such effect. Cytokine array and pathway enrichment analysis of the Mϕ conditioned media and septic sera identify TNFα (tumor necrosis factor α) as the main downstream effector of miR-146a-5p-TLR7 signaling responsible for the EC barrier dysfunction, which is further supported by neutralizing anti-TNFα antibody intervention. Together, these data demonstrate that TLR7 activation elicits pulmonary inflammation and endothelial barrier disruption by sensing extracellular miR-146a-5p and contributes to sepsis-associated acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Huang Huang
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Jing Zhu
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Lili Gu
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Jiang Hu
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Xiujing Feng
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland
| | - Sheng Wang
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Yang Yang
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Ping Cui
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Shao-Hsuan Lin
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Andrew Suen
- Center for Shock, Trauma, and Anesthesiology Research and
| | | | | | - Maureen A. Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland
| | - Yunbo Ke
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Chen-ou Zhang
- Division of Pulmonary and Critical Care Medicine, School of Medicine, and
| | - Anna A. Birukova
- Division of Pulmonary and Critical Care Medicine, School of Medicine, and
| | - Konstantin G. Birukov
- Center for Shock, Trauma, and Anesthesiology Research and
- Division of Pulmonary and Critical Care Medicine, School of Medicine, and
| | - Wei Chao
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Lin Zou
- Center for Shock, Trauma, and Anesthesiology Research and
| |
Collapse
|
29
|
Wang X, Sun Y, Wang Q, Liu F, Yang W, Sui X, Yang J, Zhang M, Wang S, Xiao Z, Luo Y, Wang Y, Zhu T. Potential Common Mechanisms of Cytotoxicity Induced by Amide Herbicides via TRPA1 Channel Activation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:7985. [PMID: 35805655 PMCID: PMC9266004 DOI: 10.3390/ijerph19137985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022]
Abstract
The “Multi-Threat Medical Countermeasure (MTMC)” strategy was proposed to develop a single drug with therapeutic efficacy against multiple pathologies or broad-spectrum protection against various toxins with common biochemical signals, molecular mediators, or cellular processes. This study demonstrated that cytotoxicity, expression of transient receptor potential cation channel subfamily A member 1 (TRPA1) mRNA, and intracellular calcium influx were increased in A549 cells exposed to amide herbicides (AHs), in which the order of cytotoxicity was metolachlor > acetochlor > propisochlor > alachlor > butachlor > propanil > pretilachlor, based on IC50 values of 430, 524, 564, 565, 619, 831, and 2333 μM, respectively. Inhibition/knockout of TRPA1 efficiently protected against cytotoxicity, decreased TRPA1 mRNA expression, and reduced calcium influx. The results suggested that the TRPA1 channel could be a key common target for AHs poisoning. The order of TRPA1 affinity for AHs was propanil > pretilachlor > metolachlor > (propiso/ala/aceto/butachlor), based on KD values of 16.2, 309, and 364 μM, respectively. The common molecular mechanisms of TRPA1-AHs interactions were clarified, including toxicity-effector groups (benzene ring, nitrogen/oxygen-containing functional groups, halogen) and residues involved in interactions (Lys787, Leu982). This work provides valuable information for the development of TRPA1 as a promising therapeutic target for broad-spectrum antitoxins.
Collapse
Affiliation(s)
- Xiaoning Wang
- Institute of Process Equipment and Environmental Engineering, School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China;
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Yangyang Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Qian Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Fengying Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Weijie Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Xin Sui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Jun Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Minmin Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Shuai Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Zhenyu Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (Y.S.); (Q.W.); (F.L.); (W.Y.); (X.S.); (J.Y.); (M.Z.); (S.W.); (Z.X.); (Y.W.)
| | - Tong Zhu
- Institute of Process Equipment and Environmental Engineering, School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China;
| |
Collapse
|
30
|
Sasso J, Ambrose BJB, Tenchov R, Datta RS, Basel MT, DeLong RK, Zhou QA. The Progress and Promise of RNA Medicine─An Arsenal of Targeted Treatments. J Med Chem 2022; 65:6975-7015. [PMID: 35533054 PMCID: PMC9115888 DOI: 10.1021/acs.jmedchem.2c00024] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Indexed: 02/08/2023]
Abstract
In the past decade, there has been a shift in research, clinical development, and commercial activity to exploit the many physiological roles of RNA for use in medicine. With the rapid success in the development of lipid-RNA nanoparticles for mRNA vaccines against COVID-19 and with several approved RNA-based drugs, RNA has catapulted to the forefront of drug research. With diverse functions beyond the role of mRNA in producing antigens or therapeutic proteins, many classes of RNA serve regulatory roles in cells and tissues. These RNAs have potential as new therapeutics, with RNA itself serving as either a drug or a target. Here, based on the CAS Content Collection, we provide a landscape view of the current state and outline trends in RNA research in medicine across time, geography, therapeutic pipelines, chemical modifications, and delivery mechanisms.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Barbara J. B. Ambrose
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Rumiana Tenchov
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Ruchira S. Datta
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Matthew T. Basel
- College
of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Robert K. DeLong
- Nanotechnology
Innovation Center Kansas State, Kansas State
University, Manhattan, Kansas 66506, United States
| | - Qiongqiong Angela Zhou
- CAS,
a division of the American Chemical Society 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
31
|
Wang Z, Chen M, Pan X, Wang L, Yin C, Lin Q, Jiang J, Zhang Y, Wan B. Knockout of GGPPS1 restrains rab37-mediated autophagy in response to ventilator-induced lung injury. Hum Cell 2022; 35:871-884. [PMID: 35334098 PMCID: PMC8948466 DOI: 10.1007/s13577-022-00692-7] [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: 01/17/2022] [Accepted: 03/11/2022] [Indexed: 12/03/2022]
Abstract
Mechanical ventilation may cause ventilator-induced lung injury (VILI) in patients requiring ventilator support. Inhibition of autophagy is an important approach to ameliorate VILI as it always enhances lung injury after exposure to various stress agents. This study aimed to further reveal the potential mechanisms underlying the effects of geranylgeranyl diphosphate synthase large subunit 1 (GGPPS1) knockout and autophagy in VILI using C57BL/6 mice with lung-specific GGPPS1 knockout that were subjected to mechanical ventilation. The results demonstrate that GGPPS1 knockout mice exhibit significantly attenuated VILI based on the histologic score, the lung wet-to-dry ratio, total protein levels, neutrophils in bronchoalveolar lavage fluid, and reduced levels of inflammatory cytokines. Importantly, the expression levels of autophagy markers were obviously decreased in GGPPS1 knockout mice compared with wild-type mice. The inhibitory effects of GGPPS1 knockout on autophagy were further confirmed by measuring the ultrastructural change of lung tissues under transmission electron microscopy. In addition, knockdown of GGPPS1 in RAW264.7 cells reduced cyclic stretch-induced inflammation and autophagy. The benefits of GGPPS1 knockout for VILI can be partially eliminated through treatment with rapamycin. Further analysis revealed that Rab37 was significantly downregulated in GGPPS1 knockout mice after mechanical ventilation, while it was highly expressed in the control group. Simultaneously, Rab37 overexpression significantly enhances autophagy in cells that are treated with cyclin stretch, including GGPPS1 knockout cells. Collectively, our results indicate that GGPPS1 knockout results in reduced expression of Rab37 proteins, further restraining autophagy and VILI.
Collapse
Affiliation(s)
- Zexu Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Meizi Chen
- Department of General Internal Medicine, The First Hospital of Chenzhou, Chenzhou, 423000, China
| | - Xia Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Li Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Cheng Yin
- Department of Clinical Laboratory, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Qiuqi Lin
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Jingjing Jiang
- Department of Rehabilitation Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Yunlei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China.
| | - Bing Wan
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China.
| |
Collapse
|
32
|
Lung function improves after delayed treatment with CNP-miR146a following acute lung injury. NANOMEDICINE: NANOTECHNOLOGY, BIOLOGY AND MEDICINE 2022; 40:102498. [PMID: 34838994 PMCID: PMC8616767 DOI: 10.1016/j.nano.2021.102498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/09/2021] [Accepted: 11/10/2021] [Indexed: 11/20/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a highly morbid pulmonary disease characterized by hypoxic respiratory failure. Its pathogenesis is characterized by unrestrained oxidative stress and inflammation, with long-term sequelae of pulmonary fibrosis and diminished lung function. Unfortunately, prior therapeutic ARDS trials have failed and therapy is limited to supportive measures. Free radical scavenging cerium oxide nanoparticles (CNP) conjugated to the anti-inflammatory microRNA-146a (miR146a), termed CNP-miR146a, have been shown to prevent acute lung injury in a pre-clinical model. In this study, we evaluated the potential of delayed treatment with CNP-miR146a at three or seven days after injury to rescue the lung from acute injury. We found that intratracheal CNP-miR146a administered three days after injury lowers pulmonary leukocyte infiltration, reduce inflammation and oxidative stress, lower pro-fibrotic gene expression and collagen deposition in the lung, and ultimately improve pulmonary function.
Collapse
|
33
|
Tarab-Ravski D, Stotsky-Oterin L, Peer D. Delivery strategies of RNA therapeutics to leukocytes. J Control Release 2022; 342:362-371. [PMID: 35041904 DOI: 10.1016/j.jconrel.2022.01.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/31/2021] [Accepted: 01/10/2022] [Indexed: 12/27/2022]
Abstract
Harnessing RNA-based therapeutics for cancer, inflammation, and viral diseases is hindered by poor delivery of therapeutic RNA molecules. Targeting leukocytes to treat these conditions holds great promise, as they are key participants in their initiation, drug response, and treatment. The various extra- and intra-cellular obstacles that impediment the clinical implementation of therapeutic RNA can be overcome by utilizing drug delivery systems. However, delivery of therapeutic RNA to leukocytes poses an even greater challenge as these cells are difficult to reach and transfect upon systemic administration. This review briefly describes the existing successful delivery strategies that efficiently target leukocytes in vivo and discuss their potential clinical applicability.
Collapse
Affiliation(s)
- Dana Tarab-Ravski
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
34
|
Riazanski V, Mauleon G, Lucas K, Walker S, Zimnicka AM, McGrath JL, Nelson DJ. Real time imaging of single extracellular vesicle pH regulation in a microfluidic cross-flow filtration platform. Commun Biol 2022; 5:13. [PMID: 35013561 PMCID: PMC8748679 DOI: 10.1038/s42003-021-02965-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022] Open
Abstract
Extracellular vesicles (EVs) are cell-derived membranous structures carrying transmembrane proteins and luminal cargo. Their complex cargo requires pH stability in EVs while traversing diverse body fluids. We used a filtration-based platform to capture and stabilize EVs based on their size and studied their pH regulation at the single EV level. Dead-end filtration facilitated EV capture in the pores of an ultrathin (100 nm thick) and nanoporous silicon nitride (NPN) membrane within a custom microfluidic device. Immobilized EVs were rapidly exposed to test solution changes driven across the backside of the membrane using tangential flow without exposing the EVs to fluid shear forces. The epithelial sodium-hydrogen exchanger, NHE1, is a ubiquitous plasma membrane protein tasked with the maintenance of cytoplasmic pH at neutrality. We show that NHE1 identified on the membrane of EVs is functional in the maintenance of pH neutrality within single vesicles. This is the first mechanistic description of EV function on the single vesicle level. Riazanski et al describe a platform to capture extracellular vesicles (EVs) using a nanoporous silicon nitride membrane, investigate the expression of NHE1 protein on the surface of EVs and monitor the transport of Na+ and H+ at the single EV level. The authors report a mechanistic function of the proteins found in EVs and specifically identify NHE1 on a single EV, where it maintains pH neutrality within single vesicles.
Collapse
Affiliation(s)
- Vladimir Riazanski
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Gerardo Mauleon
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Kilean Lucas
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Samuel Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Adriana M Zimnicka
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Deborah J Nelson
- Department of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
35
|
Zeng C, Lagier D, Lee JW, Melo MFV. Perioperative Pulmonary Atelectasis: Part I. Biology and Mechanisms. Anesthesiology 2022; 136:181-205. [PMID: 34499087 PMCID: PMC9869183 DOI: 10.1097/aln.0000000000003943] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Pulmonary atelectasis is common in the perioperative period. Physiologically, it is produced when collapsing forces derived from positive pleural pressure and surface tension overcome expanding forces from alveolar pressure and parenchymal tethering. Atelectasis impairs blood oxygenation and reduces lung compliance. It is increasingly recognized that it can also induce local tissue biologic responses, such as inflammation, local immune dysfunction, and damage of the alveolar-capillary barrier, with potential loss of lung fluid clearance, increased lung protein permeability, and susceptibility to infection, factors that can initiate or exaggerate lung injury. Mechanical ventilation of a heterogeneously aerated lung (e.g., in the presence of atelectatic lung tissue) involves biomechanical processes that may precipitate further lung damage: concentration of mechanical forces, propagation of gas-liquid interfaces, and remote overdistension. Knowledge of such pathophysiologic mechanisms of atelectasis and their consequences in the healthy and diseased lung should guide optimal clinical management.
Collapse
Affiliation(s)
- Congli Zeng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David Lagier
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jae-Woo Lee
- Department of Anesthesia, University of California San Francisco, San Francisco, CA, USA
| | - Marcos F. Vidal Melo
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Nof E, Artzy‐Schnirman A, Bhardwaj S, Sabatan H, Waisman D, Hochwald O, Gruber M, Borenstein‐Levin L, Sznitman J. Ventilation‐induced epithelial injury drives biological onset of lung trauma in vitro and is mitigated with prophylactic anti‐inflammatory therapeutics. Bioeng Transl Med 2021; 7:e10271. [PMID: 35600654 PMCID: PMC9115701 DOI: 10.1002/btm2.10271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/27/2021] [Accepted: 11/08/2021] [Indexed: 01/25/2023] Open
Abstract
Mortality rates among patients suffering from acute respiratory failure remain perplexingly high despite the maintenance of blood oxygen homeostasis during ventilatory support. The biotrauma hypothesis advocates that mechanical forces from invasive ventilation trigger immunological mediators that spread systemically. Yet, how these forces elicit an immune response remains unclear. Here, a biomimetic in vitro three‐dimensional (3D) upper airways model allows to recapitulate lung injury and immune responses induced during invasive mechanical ventilation in neonates. Under such ventilatory support, flow‐induced stresses injure the bronchial epithelium of the intubated airways model and directly modulate epithelial cell inflammatory cytokine secretion associated with pulmonary injury. Fluorescence microscopy and biochemical analyses reveal site‐specific susceptibility to epithelial erosion in airways from jet‐flow impaction and are linked to increases in cell apoptosis and modulated secretions of cytokines IL‐6, ‐8, and ‐10. In an effort to mitigate the onset of biotrauma, prophylactic pharmacological treatment with Montelukast, a leukotriene receptor antagonist, reduces apoptosis and pro‐inflammatory signaling during invasive ventilation of the in vitro model. This 3D airway platform points to a previously overlooked origin of lung injury and showcases translational opportunities in preclinical pulmonary research toward protective therapies and improved protocols for patient care.
Collapse
Affiliation(s)
- Eliram Nof
- Faculty of Biomedical Engineering Technion ‐ Israel Institute of Technology Haifa Israel
| | - Arbel Artzy‐Schnirman
- Faculty of Biomedical Engineering Technion ‐ Israel Institute of Technology Haifa Israel
| | - Saurabh Bhardwaj
- Faculty of Biomedical Engineering Technion ‐ Israel Institute of Technology Haifa Israel
| | - Hadas Sabatan
- Faculty of Biomedical Engineering Technion ‐ Israel Institute of Technology Haifa Israel
| | - Dan Waisman
- Faculty of Medicine Technion ‐ Israel Institute of Technology Haifa Israel
- Department of Neonatology Carmel Medical Center Haifa Israel
| | - Ori Hochwald
- Faculty of Medicine Technion ‐ Israel Institute of Technology Haifa Israel
- Department of Neonatology Ruth Rappaport Children's Hospital, Rambam Healthcare Haifa Israel
| | - Maayan Gruber
- Azrieli Faculty of Medicine Bar‐Ilan University Safed Israel
- Department of Otolaryngology‐Head and Neck Surgery Galilee Medical Center Nahariya Israel
| | - Liron Borenstein‐Levin
- Faculty of Medicine Technion ‐ Israel Institute of Technology Haifa Israel
- Department of Neonatology Ruth Rappaport Children's Hospital, Rambam Healthcare Haifa Israel
| | - Josué Sznitman
- Faculty of Biomedical Engineering Technion ‐ Israel Institute of Technology Haifa Israel
| |
Collapse
|
37
|
Bellio MA, Young KC, Milberg J, Santos I, Abdullah Z, Stewart D, Arango A, Chen P, Huang J, Williams K, Kelly K, Sterling S, Khan A, Xu X, Shapiro GC, Mitrani MI. Amniotic fluid-derived extracellular vesicles: characterization and therapeutic efficacy in an experimental model of bronchopulmonary dysplasia. Cytotherapy 2021; 23:1097-1107. [PMID: 34538718 DOI: 10.1016/j.jcyt.2021.07.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/12/2021] [Accepted: 07/22/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND AIMS Extracellular vesicles (EVs) are being tested for their use as novel therapeutics. However, the optimal source of EVs is currently under investigation. Amniotic fluid (AF) is a natural source of EVs that can be easily obtained for use in regenerative medicine, yet AF-EV characterization has not been fully explored. METHODS Here the authors demonstrate AF as a rich source of EVs and identify the microRNA and proteomic cargo. Bioinformatics analysis of this cargo revealed multiple pathway targets, including immunomodulatory, anti-inflammatory and free radical scavenging networks. The authors further demonstrated the therapeutic potential of this EV product as a novel preventative agent for bronchopulmonary dysplasia (BPD). RESULTS Intra-tracheal administration of AF-EVs preserved alveolar development, attenuated vascular remodeling and pulmonary hypertension, decreased lung pro-inflammatory cytokine expression and reduced macrophage infiltration in an experimental BPD model. CONCLUSIONS The authors' results suggest that AF is a viable biological fluid for EV harvest and that AF-EVs have strong therapeutic potential for pulmonary diseases, such as BPD, warranting further development to transition this novel EV product into the clinic.
Collapse
Affiliation(s)
| | - Karen C Young
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Julian Milberg
- Organicell Regenerative Medicine, Inc, Miami, Florida, USA
| | - Ivan Santos
- Organicell Regenerative Medicine, Inc, Miami, Florida, USA
| | - Zanub Abdullah
- Organicell Regenerative Medicine, Inc, Miami, Florida, USA
| | | | - Alissa Arango
- Organicell Regenerative Medicine, Inc, Miami, Florida, USA
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kevin Williams
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kaitlyn Kelly
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shanique Sterling
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Xiumin Xu
- AssureImmune LLC, Miami, Florida, USA
| | | | | |
Collapse
|
38
|
Barutta F, Corbetta B, Bellini S, Guarrera S, Matullo G, Scandella M, Schalkwijk C, Stehouwer CD, Chaturvedi N, Soedamah-Muthu SS, Durazzo M, Gruden G. MicroRNA 146a is associated with diabetic complications in type 1 diabetic patients from the EURODIAB PCS. J Transl Med 2021; 19:475. [PMID: 34823560 PMCID: PMC8614036 DOI: 10.1186/s12967-021-03142-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/11/2021] [Indexed: 12/11/2022] Open
Abstract
Background MicroRNA-146a-5p (miR-146a-5p) is a key regulator of inflammatory processes. Expression of miR-146a-5p is altered in target organs of diabetic complications and deficiency of miR-146a-5p has been implicated in their pathogenesis. We investigated if serum miR-146a-5p levels were independently associated with micro/macrovascular complications of type 1 diabetes (DM1). Methods A nested case–control study from the EURODIAB PCS of 447 DM1 patients was performed. Cases (n = 294) had one or more complications of diabetes, whereas controls (n = 153) did not have any complication. Total RNA was isolated from all subjects and miR-146a-5p levels measured by qPCR. Both the endogenous controls U6 snRNA and the spike (Cel-miR-39) were used to normalize the results. Logistic regression analysis was carried out to investigate the association of miR-146a-5p with diabetes complications. Results MiR-146a-5p levels were significantly lower in cases [1.15 (0.32–3.34)] compared to controls [1.74 (0.44–6.74) P = 0.039]. Logistic regression analysis showed that levels of miR-146a-5p in the upper quartile were inversely associated with reduced odds ratio (OR) of all complications (OR 0.34 [95% CI 0.14–0.76]) and particularly with cardiovascular diseases (CVD) (OR 0.31 [95% CI 0.11–0.84]) and diabetic retinopathy (OR 0.40 [95% CI 0.16–0.99]), independently of age, sex, diabetes duration, A1c, hypertension, AER, eGFR, NT-proBNP, and TNF-α. Conclusions In this large cohort of DM1 patients, we reported an inverse and independent association of miR-146a-5p with diabetes chronic complications and in particular with CVD and retinopathy, suggesting that miR-146a-5p may be a novel candidate biomarker of DM1 complications. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03142-4.
Collapse
Affiliation(s)
- Federica Barutta
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy.
| | - Beatrice Corbetta
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| | - Stefania Bellini
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| | - Simonetta Guarrera
- Italian Institute for Genomic Medicine, IIGM, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Giuseppe Matullo
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy.,Medical Genetics Unit, AOU Città Della Salute E Della Scienza, Turin, Italy
| | - Michela Scandella
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| | - Casper Schalkwijk
- Department of Internal Medicine and Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Coen D Stehouwer
- Department of Internal Medicine and Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Nish Chaturvedi
- Institute of Cardiovascular Science, University College London, London, UK
| | - Sabita S Soedamah-Muthu
- Center of Research On Psychology in Somatic Diseases (CORPS), Department of Medical and Clinical Psychology, Tilburg University, Tilburg, The Netherlands.,Institute for Food, Nutrition and Health, University of Reading, Reading, UK
| | - Marilena Durazzo
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| | - Gabriella Gruden
- Diabetic Nephropathy Laboratory, Department of Medical Sciences, University of Turin, C/so Dogliotti 14, 10126, Turin, Italy
| |
Collapse
|
39
|
Mallampalli RK, Adair J, Elhance A, Farkas D, Chafin L, Long ME, De M, Mora AL, Rojas M, Peters V, Bednash JS, Tsai M, Londino JD. Interferon Lambda Signaling in Macrophages Is Necessary for the Antiviral Response to Influenza. Front Immunol 2021; 12:735576. [PMID: 34899695 PMCID: PMC8655102 DOI: 10.3389/fimmu.2021.735576] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/04/2021] [Indexed: 01/14/2023] Open
Abstract
Interferon lambda (IFNλ) signaling is a promising therapeutic target against viral infection in murine models, yet little is known about its molecular regulation and its cognate receptor, interferon lambda receptor 1 (IFNLR1) in human lung. We hypothesized that the IFNλ signaling axis was active in human lung macrophages. In human alveolar macrophages (HAMs), we observed increased IFNLR1 expression and robust increase in interferon-stimulated gene (ISG) expression in response to IFNλ ligand. While human monocytes express minimal IFNLR1, differentiation of monocytes into macrophages with macrophage colony-stimulating factor (M-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF) increased IFNLR1 mRNA, IFNLR1 protein expression, and cellular response to IFNλ ligation. Conversely, in mice, M-CSF or GM-CSF stimulated macrophages failed to produce ISGs in response to related ligands, IFNL2 or IFNL3, suggesting that IFNLR1 signaling in macrophages is species-specific. We next hypothesized that IFNλ signaling was critical in influenza antiviral responses. In primary human airway epithelial cells and precision-cut human lung slices, influenza infection substantially increased IFNλ levels. Pretreatment of both HAMs and differentiated human monocytes with IFNL1 significantly inhibited influenza infection. IFNLR1 knockout in the myeloid cell line, THP-1, exhibited reduced interferon responses to either direct or indirect exposure to influenza infection suggesting the indispensability of IFNLR1 for antiviral responses. These data demonstrate the presence of IFNλ - IFNLR1 signaling axis in human lung macrophages and a critical role of IFNλ signaling in combating influenza infection.
Collapse
Affiliation(s)
- Rama K. Mallampalli
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Jessica Adair
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Ajit Elhance
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Daniela Farkas
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Lexie Chafin
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Matthew E. Long
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States,Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Mithu De
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Ana L. Mora
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Mauricio Rojas
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Victor Peters
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Joseph S. Bednash
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - MuChun Tsai
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - James D. Londino
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Davis Heart and Lung Research Institute, Columbus, Ohio, United States,*Correspondence: James D. Londino,
| |
Collapse
|
40
|
Olivieri F, Prattichizzo F, Giuliani A, Matacchione G, Rippo MR, Sabbatinelli J, Bonafè M. miR-21 and miR-146a: The microRNAs of inflammaging and age-related diseases. Ageing Res Rev 2021; 70:101374. [PMID: 34082077 DOI: 10.1016/j.arr.2021.101374] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
The first paper on "inflammaging" published in 2001 paved the way for a unifying theory on how and why aging turns out to be the main risk factor for the development of the most common age-related diseases (ARDs). The most exciting challenge on this topic was explaining how systemic inflammation steeps up with age and why it shows different rates among individuals of the same chronological age. The "epigenetic revolution" in the past twenty years conveyed that the assessment of the individual genetic make-up is not enough to depict the trajectories of age-related inflammation. Accordingly, others and we have been focusing on the role of non-coding RNA, i.e. microRNAs (miRNAs), in inflammaging. The results obtained in the latest 10 years underpinned the key role of a miRNA subset that we have called inflammamiRs, owing to their ability to master (NF-κB)-driven inflammatory pathways. In this review, we will focus on two inflammamiRs, i.e. miR-21-5p and miR-146a-5p, which target a variety of molecules belonging to the NF-κB/NLRP3 pathways. The interplay between miR-146a-5p and IL-6 in the context of aging and ARDs will also be highlighted. We will also provide the most relevant evidence suggesting that circulating inflammamiRs, along with IL-6, can measure the degree of inflammaging.
Collapse
|
41
|
Fattal E, Fay F. Nanomedicine-based delivery strategies for nucleic acid gene inhibitors in inflammatory diseases. Adv Drug Deliv Rev 2021; 175:113809. [PMID: 34033819 DOI: 10.1016/j.addr.2021.05.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 02/07/2023]
Abstract
Thanks to their abilities to modulate the expression of virtually any genes, RNA therapeutics have attracted considerable research efforts. Among the strategies focusing on nucleic acid gene inhibitors, antisense oligonucleotides and small interfering RNAs have reached advanced clinical trial phases with several of them having recently been marketed. These successes were obtained by overcoming stability and cellular delivery issues using either chemically modified nucleic acids or nanoparticles. As nucleic acid gene inhibitors are promising strategies to treat inflammatory diseases, this review focuses on the barriers, from manufacturing issues to cellular/subcellular delivery, that still need to be overcome to deliver the nucleic acids to sites of inflammation other than the liver. Furthermore, key examples of applications in rheumatoid arthritis, inflammatory bowel, and lung diseases are presented as case studies of systemic, oral, and lung nucleic acid delivery.
Collapse
|
42
|
A Peptide Inhibitor of Peroxiredoxin 6 Phospholipase A 2 Activity Significantly Protects against Lung Injury in a Mouse Model of Ventilator Induced Lung Injury (VILI). Antioxidants (Basel) 2021; 10:antiox10060925. [PMID: 34200443 PMCID: PMC8226847 DOI: 10.3390/antiox10060925] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Ventilator induced lung injury (VILI) is a lung injury syndrome associated with mechanical ventilation, most frequently for treatment of Acute Lung Injury (ALI), and generally secondary to the use of greater than physiologic tidal volumes. To reproduce this syndrome experimentally, C57Bl/6 mice were intubated and ventilated with low (4 mL/Kg body weight) or high (12 mL/Kg) tidal volume for 6 h. Lung parameters with low volume ventilation were unchanged from non-ventilated (control) mice. High tidal volume ventilation resulted in marked lung injury with increased neutrophils in the bronchoalveolar lavage fluid (BALf) indicating lung inflammation, increase in both protein in BALf and lung dry/wet weight indicating lung edema, increased lung thiobarbituric acid reactive substances (TBARS) and 8-isoprostanes indicating lung lipid peroxidation, and increased lung protein carbonyls indicating protein oxidation. Either intratracheal or intravenous pretreatment of mice with a 9 amino acid peptide called peroxiredoxin 6 inhibitor peptide-2 (PIP-2) significantly reduced all parameters of lung injury by ~50–80%. PIP-2 inhibits NADPH oxidase type 2 (NOX2) activation. We propose that PIP-2 does not affect the mechanically induced lung damage component of VILI but does significantly reduce the secondary inflammatory component.
Collapse
|