1
|
Zhang Y, Wu K, Mao D, Iberg CA, Yin-Declue H, Sun K, Wikfors HA, Keeler SP, Li M, Young D, Yantis J, Crouch EC, Chartock JR, Han Z, Byers DE, Brody SL, Romero AG, Holtzman MJ. A first-in-kind MAPK13 inhibitor that can correct stem cell reprogramming and post-injury disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608990. [PMID: 39229202 PMCID: PMC11370402 DOI: 10.1101/2024.08.21.608990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The stress kinase MAPK13 (aka p38δ-MAPK) is an attractive entry point for therapeutic intervention because it regulates the structural remodeling that can develop after epithelial barrier injury in the lung and likely other tissue sites. However, a selective, safe, and effective MAPK13 inhibitor is not yet available for experimental or clinical application. Here we identify a first-in-kind MAPK13 inhibitor using structure-based drug design combined with a screening funnel for cell safety and molecular specificity. This inhibitor (designated NuP-4) down-regulates basal-epithelial stem cell reprogramming, structural remodeling, and pathophysiology equivalently to Mapk13 gene-knockout in mouse and mouse organoid models of post-viral lung disease. This therapeutic benefit persists after stopping treatment as a sign of disease modification and attenuates key aspects of inflammation and remodeling as an indication of disease reversal. Similarly, NuP-4 treatment can directly control cytokine-stimulated growth, immune activation, and mucinous differentiation in human basal-cell organoids. The data thereby provide a new tool and potential fix for long-term stem cell reprogramming after viral injury and related conditions that require MAPK13 induction-activation.
Collapse
Affiliation(s)
- Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Courtney A Iberg
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Huiqing Yin-Declue
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Hallie A Wikfors
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Ming Li
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Deanna Young
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Erika C Crouch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Joshua R Chartock
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Zhenfu Han
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L Brody
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Arthur G Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
- NuPeak Therapeutics Inc., St. Louis, MO 63105
| |
Collapse
|
2
|
Wu K, Zhang Y, Mao D, Iberg CA, Yin-Declue H, Sun K, Keeler SP, Wikfors HA, Young D, Yantis J, Austin SR, Byers DE, Brody SL, Crouch EC, Romero AG, Holtzman MJ. MAPK13 controls structural remodeling and disease after epithelial injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596863. [PMID: 38895360 PMCID: PMC11185504 DOI: 10.1101/2024.05.31.596863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
All living organisms are charged with repair after injury particularly at epithelial barrier sites, but in some cases this response leads instead to structural remodeling and long-term disease. Identifying the molecular and cellular control of this divergence is key to disease modification. In that regard, stress kinase control of epithelial stem cells is a rational entry point for study. Here we examine the potential for mitogen-activated protein kinase 13 (MAPK13) regulation of epithelial stem cells using models of respiratory viral injury and post-viral lung disease. We show that Mapk13 gene-knockout mice handle acute infectious illness as expected but are protected against structural remodeling manifest as basal-epithelial stem cell (basal-ESC) hyperplasia-metaplasia, immune activation, and mucinous differentiation. In corresponding cell models, Mapk13-deficiency directly attenuates basal-ESC growth and organoid formation. Extension to human studies shows marked induction/activation of basal-cell MAPK13 in clinical samples of comparable remodeling found in asthma and COPD. Here again, MAPK13 gene-knockdown inhibits human basal-ESC growth in culture. Together, the data identify MAPK13 as a control for structural remodeling and disease after epithelial injury and as a suitable target for down-regulation as a disease-modifying strategy.
Collapse
Affiliation(s)
- Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Courtney A. Iberg
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Huiqing Yin-Declue
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelly Sun
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P. Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Hallie A. Wikfors
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Deanna Young
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen R. Austin
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L. Brody
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Erika C. Crouch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Arthur G. Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J. Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
- NuPeak Therapeutics Inc., St. Louis, MO 63105
| |
Collapse
|
3
|
Saleem S. Targeting MAPK signaling: A promising approach for treating inflammatory lung disease. Pathol Res Pract 2024; 254:155122. [PMID: 38246034 DOI: 10.1016/j.prp.2024.155122] [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: 09/17/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
The extracellular signals that initiate intracellular reactions are dispatched by the mitogen-activated protein kinases (MAPKs), which oversee a multitude of cellular activities. p38, Extracellular signal-regulated kinase (ERK), and c-Jun NH2-terminal kinase (JNK) are members of the vertebrate family of MAPKs, and each MAPK signaling pathway consists of a MAPK kinase (MAP3K), a MAPK kinase (MAP2K), and a MAPK. These signaling pathways orchestrate numerous cellular processes, including cell growth, survival, differentiation, and apoptosis. The emergence of various inflammatory respiratory diseases in humans has been linked to the dysregulation of MAPK signaling pathways. Conditions such as asthma, lung cancer, pulmonary fibrosis, and COPD are among the prevalent respiratory ailments where MAPK plays a pivotal role. Additionally, MAPK is implicated in infectious diseases, including COVID-19, pneumonia, and tuberculosis. COPD, asthma, emphysema, chronic bronchitis, and other inflammatory lung disorders highlight the significance of MAPK as a potential target for therapeutic development. Further studies are needed to delve into the molecular mechanisms by which the MAPK signaling pathway contributes to inflammatory lung disorders, representing an area that demands continued research.
Collapse
Affiliation(s)
- Shakir Saleem
- Department of Public Health, College of Health Sciences, Saudi Electronic University, P.O. Box 93499, Riyadh 11673, Saudi Arabia.
| |
Collapse
|
4
|
Holtzman MJ, Zhang Y, Wu K, Romero AG. Mitogen-activated protein kinase-guided drug discovery for post-viral and related types of lung disease. Eur Respir Rev 2024; 33:230220. [PMID: 38417971 PMCID: PMC10900067 DOI: 10.1183/16000617.0220-2023] [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: 10/27/2023] [Accepted: 01/18/2024] [Indexed: 03/01/2024] Open
Abstract
Respiratory viral infections are a major public health problem, with much of their morbidity and mortality due to post-viral lung diseases that progress and persist after the active infection is cleared. This paradigm is implicated in the most common forms of chronic lung disease, such as asthma and COPD, as well as other virus-linked diseases including progressive and long-term coronavirus disease 2019. Despite the impact of these diseases, there is a lack of small-molecule drugs available that can precisely modify this type of disease process. Here we will review current progress in understanding the pathogenesis of post-viral and related lung disease with characteristic remodelling phenotypes. We will also develop how this data leads to mitogen-activated protein kinase (MAPK) in general and MAPK13 in particular as key druggable targets in this pathway. We will also explore recent advances and predict the future breakthroughs in structure-based drug design that will provide new MAPK inhibitors as drug candidates for clinical applications. Each of these developments point to a more effective approach to treating the distinct epithelial and immune cell based mechanisms, which better account for the morbidity and mortality of post-viral and related types of lung disease. This progress is vital given the growing prevalence of respiratory viruses and other inhaled agents that trigger stereotyped progression to acute illness and chronic disease.
Collapse
Affiliation(s)
- Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- NuPeak Therapeutics Inc., St. Louis, MO, USA
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Arthur G Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
5
|
Liu X, Zhao L, Wang R, Tang Z. TRPM2 exacerbates airway inflammation by regulating oxidized-CaMKⅡ in allergic asthma. Heliyon 2024; 10:e23634. [PMID: 38187281 PMCID: PMC10767383 DOI: 10.1016/j.heliyon.2023.e23634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Background Airway epithelial cells play important roles in allergic asthma. Transient receptor potential melastatin-related 2 (TRPM2) and oxidized Ca2+/calmodulin-dependent protein kinase Ⅱ (ox-CaMKⅡ) participate in the airway inflammation. This study aimed to analyze the effects of TRPM2 on ox-CaMKⅡ in the airway epithelial cells during allergic asthma. Methods BEAS-2B cells were treated with different dose of IL-13 (0, 5, 10, 20 ng/mL) for 24 h to analyze the changes of TRPM2 and ox-CaMKⅡ protein. Cells expressing different level of TRPM2 were obtained by transfection of TRPM2 siRNA or TRPM2-short cDNA. The transfected cells were treated with 10 ng/mL of IL-13 to analyze the effects of TRPM2 on the ox-CaMKⅡ. A CaMKⅡ inhibitor KN-93 was used to confirm the effects of TRPM2 on levels of ox-CaMKⅡ, p-MEK and p-ERK in the IL-13-treated BEAS-2B cells. Wild-type (WT) mice and TRPM2-knockout (TRPM2-/-) mice were induced by ovalbumin (OVA) to compare the differences of inflammation, levels of ox-CaMKII, p-MEK and p-ERK in airways. Results Cell viability was clearly decreased by the 20 ng/mL of IL-13. The levels of TRPM2 and ox-CaMKII protein in cells were increased with increasing doses of IL-13. Transfection of TRPM2 siRNA or TRPM2-short cDNA respectively decreased or increased the levels of ox-CaMKⅡ in the IL-13-stimulated cells. The results of KN-93 treatment were similar to the results of TRPM2 siRNA transfection, that the levels of ox-CaMKⅡ, p-MEK and p-ERK were significantly decreased in the IL-13-treated cells. Compared with the OVA-induced WT mice, levels of inflammation, ox-CaMKⅡ, p-MEK and p-ERK in the airways were significantly weakened in the OVA-induced TRPM2-/- mice. Conclusions TRPM2 plays a vital role in regulating ox-CaMKⅡ in airway epithelial cells during allergic asthma.
Collapse
Affiliation(s)
- Xueping Liu
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, 264000, China
| | - Lingyan Zhao
- Department of Nursing, Yantai Yuhuangding Hospital, 264000, China
| | - Rui Wang
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, 264000, China
| | - Zhaoying Tang
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, 264000, China
| |
Collapse
|
6
|
Fang Y, Cai J, Ren M, Zhong T, Wang D, Zhang K. Inhalable Bottlebrush Polymer Bioconjugates as Vectors for Efficient Pulmonary Delivery of Oligonucleotides. ACS NANO 2024; 18:592-599. [PMID: 38147573 PMCID: PMC10786149 DOI: 10.1021/acsnano.3c08660] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Antisense oligonucleotides hold therapeutic promise for various lung disorders, but their efficacy is limited by suboptimal delivery. To address this challenge, we explored the use of inhaled bottlebrush polymer-DNA conjugates, named pacDNA, as a delivery strategy. Inhaled pacDNA exhibits superior mucus penetration, achieving a uniform and sustained lung distribution in mice. Targeting the 5' splice site of an aberrant enhanced green fluorescence protein (EGFP) pre-mRNA in EGFP-654 mice, inhaled pacDNA more efficiently corrects splicing than a B-peptide conjugate and restores EGFP expression in the lung. Additionally, in an orthotopic NCI-H358 non-small-cell lung tumor mouse model, inhaled pacDNA targeting wild-type KRAS mRNA effectively suppresses KRAS expression and inhibits lung tumor growth, requiring a substantially lower dosage compared to intravenously injected pacDNA. These findings demonstrate the potential of bottlebrush polymer-DNA conjugates as a promising agent for enhanced oligonucleotide therapy in the lung and advancing the treatment landscape for lung disorders.
Collapse
Affiliation(s)
- Yang Fang
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Jiansong Cai
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Mengqi Ren
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Tongtong Zhong
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Dali Wang
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Ke Zhang
- Department of Chemistry and Chemical
Biology, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
7
|
Keeler SP, Wu K, Zhang Y, Mao D, Li M, Iberg CA, Austin SR, Glaser SA, Yantis J, Podgorny S, Brody SL, Chartock JR, Han Z, Byers DE, Romero AG, Holtzman MJ. A potent MAPK13-14 inhibitor prevents airway inflammation and mucus production. Am J Physiol Lung Cell Mol Physiol 2023; 325:L726-L740. [PMID: 37847710 PMCID: PMC11068410 DOI: 10.1152/ajplung.00183.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023] Open
Abstract
Common respiratory diseases continue to represent a major public health problem, and much of the morbidity and mortality is due to airway inflammation and mucus production. Previous studies indicated a role for mitogen-activated protein kinase 14 (MAPK14) in this type of disease, but clinical trials are unsuccessful to date. Our previous work identified a related but distinct kinase known as MAPK13 that is activated in respiratory airway diseases and is required for mucus production in human cell-culture models. Support for MAPK13 function in these models came from effectiveness of MAPK13 versus MAPK14 gene-knockdown and from first-generation MAPK13-14 inhibitors. However, these first-generation inhibitors were incompletely optimized for blocking activity and were untested in vivo. Here we report the next generation and selection of a potent MAPK13-14 inhibitor (designated NuP-3) that more effectively downregulates type-2 cytokine-stimulated mucus production in air-liquid interface and organoid cultures of human airway epithelial cells. We also show that NuP-3 treatment prevents respiratory airway inflammation and mucus production in new minipig models of airway disease triggered by type-2 cytokine challenge or respiratory viral infection. The results thereby provide the next advance in developing a small-molecule kinase inhibitor to address key features of respiratory disease.NEW & NOTEWORTHY This study describes the discovery of a potent mitogen-activated protein kinase 13-14 (MAPK13-14) inhibitor and its effectiveness in models of respiratory airway disease. The findings thereby provide a scheme for pathogenesis and therapy of lung diseases [e.g., asthma, chronic obstructive pulmonary disease (COPD), Covid-19, postviral, and allergic respiratory disease] and related conditions that implicate MAPK13-14 function. The findings also refine a hypothesis for epithelial and immune cell functions in respiratory disease that features MAPK13 as a possible component of this disease process.
Collapse
Affiliation(s)
- Shamus P Keeler
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Kangyun Wu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Yong Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Dailing Mao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Ming Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Courtney A Iberg
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | | | - Samuel A Glaser
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jennifer Yantis
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Stephanie Podgorny
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Steven L Brody
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Joshua R Chartock
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Zhenfu Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Derek E Byers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Arthur G Romero
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Michael J Holtzman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States
- NuPeak Therapeutics Inc., St. Louis, Missouri, United States
| |
Collapse
|
8
|
Keeler SP, Wu K, Zhang Y, Mao D, Li M, Iberg CA, Austin SR, Glaser SA, Yantis J, Podgorny S, Brody SL, Chartock JR, Han Z, Byers DE, Romero AG, Holtzman MJ. A potent MAPK13-14 inhibitor prevents airway inflammation and mucus production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542451. [PMID: 37292761 PMCID: PMC10246002 DOI: 10.1101/2023.05.26.542451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Common respiratory diseases continue to represent a major public health problem, and much of the morbidity and mortality is due to airway inflammation and mucus production. Previous studies indicated a role for mitogen-activated protein kinase 14 (MAPK14) in this type of disease, but clinical trials are unsuccessful to date. Our previous work identified a related but distinct kinase known as MAPK13 that is activated in respiratory airway diseases and is required for mucus production in human cell-culture models. Support for MAPK13 function in these models came from effectiveness of MAPK13 versus MAPK14 gene-knockdown and from first-generation MAPK13-14 inhibitors. However, these first-generation inhibitors were incompletely optimized for blocking activity and were untested in vivo. Here we report the next generation and selection of a potent MAPK13-14 inhibitor (designated NuP-3) that more effectively down-regulates type-2 cytokine-stimulated mucus production in air-liquid interface and organoid cultures of human airway epithelial cells. We also show that NuP-3 treatment prevents respiratory airway inflammation and mucus production in new minipig models of airway disease triggered by type-2 cytokine challenge or respiratory viral infection. The results thereby provide the next advance in developing a small-molecule kinase inhibitor to address key features of respiratory disease.
Collapse
Affiliation(s)
- Shamus P. Keeler
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Ming Li
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Courtney A. Iberg
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | | | - Samuel A. Glaser
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer Yantis
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephanie Podgorny
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Steven L. Brody
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Joshua R. Chartock
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Zhenfu Han
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E. Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Arthur G. Romero
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J. Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
- NuPeak Therapeutics Inc., St. Louis, MO 63105
| |
Collapse
|
9
|
Makled MN, El-Sheakh AR. Fingolimod attenuates ovalbumin-induced airway inflammation via inhibiting MAPK/ERK signaling in mice. J Biochem Mol Toxicol 2023; 37:e23266. [PMID: 36468814 DOI: 10.1002/jbt.23266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/06/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
The current study was designed to investigate the potential anti-inflammatory and antioxidant effects of fingolimod against Ovalbumin (Ova)-induced allergic airway inflammation compared to dexamethasone. Fingolimod was given (0.5 mg/kg/day, p.o.) for sensitized mice 1 h before Ova challenge from Days 19 to 24. Fingolimod significantly inhibited Ova-induced elevation of inflammatory cells and eosinophils numbers in bronchoalveolar lavage fluid (BALF) and reduced concentrations of immunoglobulin E in serum and of sphingosine-1-phosphate, interleukin (IL)-4, and IL-13 in BALF. Fingolimod inhibited microvascular leakage and edema as reflected by the decreased lung/body weight index. These findings were supported by histopathological examination results showing that fingolimod substantially decreased perivascular edema and inflammatory cell infiltration. Fingolimod also attenuated Ova-induced oxidative stress as evidenced by decreased malondialdehyde concentration along with increasing concentrations of reduced glutathione and superoxide dismutase in lung tissues. Fingolimod also significantly decreased monocyte chemoattractant protein-1 (MCP-1), p-ERK, and p-P38 in lung tissues of Ova-challenged mice. In conclusion, the current study demonstrated the anti-inflammatory and antioxidant effects of fingolimod in allergic airway inflammation that might be associated with the downregulation of mitogen activated kinases signaling to decrease T helper 2 cytokine secretion (IL-4 and IL-13) and MCP-1 expression, along with the inhibition of oxidative stress.
Collapse
Affiliation(s)
- Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed R El-Sheakh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.,Future Studies and Risks Management & National Committee of Drugs, Academy of Scientific Research, Ministry of Higher Education, ElSayeda Zeinab, Egypt
| |
Collapse
|
10
|
Ceramide Nanoliposomes as Potential Therapeutic Reagents for Asthma. Cells 2023; 12:cells12040591. [PMID: 36831258 PMCID: PMC9954069 DOI: 10.3390/cells12040591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Ceramides are an emerging class of anti-inflammatory lipids, and nanoscale ceramide-delivery systems are potential therapeutic strategies for inflammatory diseases. This study investigated the therapeutic effects of ceramide nanoliposomes (CNL) on type 2 inflammation-based asthma, induced by repeated ovalbumin (OVA) challenges. Asthmatic mice intratracheally treated with ceramide-free liposomes (Ghost) displayed typical airway remodeling including mucosal accumulation and subepithelial fibrosis, whereas, in CNL-treated mice, the degree of airway remodeling was significantly decreased. Compared to the Ghost group, CNL treatment unexpectedly failed to significantly influence formation of type 2 cytokines, including IL-5 and IL-13, known to facilitate pathogenic production of airway mucus predominantly comprising MUC5AC mucin. Interestingly, CNL treatment suppressed OVA-evoked hyperplasia of MUC5AC-generating goblet cells in the airways. This suggests that CNL suppressed goblet cell hyperplasia and airway mucosal accumulation independently of type 2 cytokine formation. Mechanistically, CNL treatment suppressed cell growth and EGF-induced activation of Akt, but not ERK1/2, in a human lung epithelial cell culture system recapitulating airway goblet cell hyperplasia. Taken together, CNL is suggested to have therapeutic effects on airway remodeling in allergic asthma by targeting goblet cell hyperplasia. These findings raise the potential of ceramide-based therapies for airway diseases, such as asthma.
Collapse
|
11
|
Yu H, Su X, Lei T, Zhang L, Feng Z, Zhang C, Zhang M, Wang Y, Chen X, Liu J. Safety and efficacy of p38 mitogen-activated protein kinase inhibitors (MAPKIs) in COPD. Front Pharmacol 2022; 13:950035. [PMID: 36249771 PMCID: PMC9554617 DOI: 10.3389/fphar.2022.950035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Chronic inflammation is the core mechanism of the development of chronic obstructive pulmonary disease (COPD). Corticosteroid resistance in COPD limits its anti-inflammatory potency. p38 MAPKIs were suggested as an alternative to corticosteroids despite the fact that there is currently no systematic review evaluating existing evidence.Methods: This randomized controlled trials (RCT)-based systematic review with meta-analysis was conducted following the PRISMA statement. RCTs were searched and screened from 8 databases. Three types of data, including basic information of included studies, pre-defined outcome data, and quality assessment information were extracted. Pooling values and associated 95 % confidence intervals were deemed as statistically significant only when two-tailed p values were smaller than 0.05.Results: This study included 10 RCTs with a total population of 1,751 [age, mean (SD) = 64.39 (8.06)]. Safety and several efficacy indicators of lung function, inflammatory biomarkers, and quality of life were meta-analyzed. Despite the improvement of post-bronchodilator-forced vital capacity (FVC), no difference between p38 MAPKIs and placebo was found in both safety and efficacy.Conclusion: Compared with placebo, p38 MAPKIs are safe but did not show any significant effects in the COPD population. Results of this study should be regarded with caution due to the small number of included studies and heterogeneity from combining different p38 MAPKIs as a whole.Systematic Review registration: PROSPERO #CRD42022302890.
Collapse
Affiliation(s)
- Haichuan Yu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaojie Su
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ting Lei
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lu Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhouzhou Feng
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Chuchu Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Meng Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yalei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xinlong Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Intensive Care Unit, The First Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Jian Liu,
| |
Collapse
|
12
|
rhCNB Improves Cyclophosphamide-Induced Immunodeficiency in BALB/c Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4891399. [PMID: 36204132 PMCID: PMC9532092 DOI: 10.1155/2022/4891399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 12/03/2022]
Abstract
Background This study aims to explore the immunomodulatory effect of rhCNB on mice with cyclophosphamide (CTX)-induced immunodeficiency through TLR4/MAPK pathway. Methods BALB/c mice were randomly divided into three groups: a negative control group, an immunosuppression model group, and a rhCNB treatment group. Tail vein injection of cyclophosphamide (40 mg/kg) was used to establish a mouse immunosuppression model. Intraperitoneal injection of rhCNB (20 mg/kg) was administered to the treatment group, whereas equal quantities of normal saline were given to the control group and model group. Perform peripheral blood routine of CD4, CD8, and CD19 lymphocyte subsets and peripheral blood Th1/Th2 cell subsets 24 hours after the last administration. RT-PCR was used to detect mRNA levels of TLR4, P38, JNK, T-bet, and GATA3, the spleen immune organ index was measured, and the histopathological status of the spleen and thymus was observed. Results The results showed that compared with the control group, WBC, PLT, LYM, NEU, immune organ index, CD4+/CD8+ and CD19+ subgroup ratio, and peripheral blood Th1/Th2 cell subgroups decreased in the model group. The mRNA levels of TLR4, P38, JNK, T-bet, and GATA3 decreased compared with the model group, while they increased in the treatment group. Conclusions rhCNB has an immunomodulatory effect by regulating the expression of Th1/Th2 cytokine balance through the TLR4/MAPK signaling pathway and promoting the differentiation and proliferation of lymphocytes, thereby improving the immune function.
Collapse
|
13
|
Protective Effects of Platycodin D3 on Airway Remodeling and Inflammation via Modulating MAPK/NF-κB Signaling Pathway in Asthma Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1612829. [PMID: 35990822 PMCID: PMC9385299 DOI: 10.1155/2022/1612829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/06/2022] [Accepted: 07/06/2022] [Indexed: 11/20/2022]
Abstract
Background Asthma is a disease with airway hyperresponsive and airway inflammation. Platycodin D is a triterpenoid saponin extracted from Platycodon grandiflorus root, which has various pharmacological activities. The study mainly explored the effects of platycodin D3 (PD3) in airway remodeling and inflammation of asthma. Methods The ovalbumin (OVA)-induced asthma mice were given PD3 (20 mg/kg, 40 mg/kg, and 80 mg/kg) in different groups. The asthma mice administrated with dexamethasone (DXM) were enrolled as the positive control group, and the normal control mice and asthma model mice separately received the same volume of saline. Mouse airway lung dynamic compliance (Cdyn) and total airway resistance (RL) were measured by the EMKA animal lung function analysis system. The inflammation factor levels were estimated by ELISA. Histopathological changes were tested by HE and PAS staining. The protein and phosphorylation levels of NF-κBp65, p38, ERK1/2, and JNK1/2 were detected by Western blot. Results In asthmatic mice, PD3 enhanced the airway Cdyn and decreased RL to improve the airway hyperreactivity and alleviated the pathological injury of lung tissues. In addition, PD3 could reduce the infiltration of inflammatory cells in BALF and suppress the levels of eotaxin, IL-4, IL-5, IL-13, IFN-γ, and IgE. Furthermore, PD3 treatment inhibited the phosphorylation of NF-κBp65, p38, ERK1/2, and JNK1/2 proteins in asthma mice. Conclusion PD3 treatment alleviated the airway remodeling and inflammation in asthmatic mice, which might be related to downregulating the phosphorylated proteins in the MAPK/NF-κB signaling pathway.
Collapse
|
14
|
Armani E, Capaldi C, Bagnacani V, Saccani F, Aquino G, Puccini P, Facchinetti F, Martucci C, Moretto N, Villetti G, Patacchini R, Civelli M, Hurley C, Jennings A, Alcaraz L, Bloomfield D, Briggs M, Daly S, Panchal T, Russell V, Wicks S, Finch H, Fitzgerald M, Fox C, Delcanale M. Design, Synthesis, and Biological Characterization of Inhaled p38α/β MAPK Inhibitors for the Treatment of Lung Inflammatory Diseases. J Med Chem 2022; 65:7170-7192. [PMID: 35546685 DOI: 10.1021/acs.jmedchem.2c00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The identification of novel inhaled p38α/β mitogen-activated protein kinases (MAPK) (MAPK14/11) inhibitors suitable for the treatment of pulmonary inflammatory conditions has been described. A rational drug design approach started from the identification of a novel tetrahydronaphthalene series, characterized by nanomolar inhibition of p38α with selectivity over p38γ and p38δ isoforms. SAR optimization of 1c is outlined, where improvements in potency against p38α and ligand-enzyme dissociation kinetics led to several compounds showing pronounced anti-inflammatory effects in vitro (inhibition of TNFα release). Targeting of the defined physicochemical properties allowed the identification of compounds 3h, 4e, and 4f, which showed, upon intratracheal instillation, low plasma levels, prolonged lung retention, and anti-inflammatory effects in a rat acute model of a bacterial endotoxin-induced pulmonary inflammation. Compound 4e, in particular, displayed remarkable efficacy and duration of action and was selected for progression in disease models of asthma and chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Elisabetta Armani
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Carmelida Capaldi
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Valentina Bagnacani
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Francesca Saccani
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Giancarlo Aquino
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paola Puccini
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Fabrizio Facchinetti
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Cataldo Martucci
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Nadia Moretto
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Gino Villetti
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Riccardo Patacchini
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Maurizio Civelli
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Chris Hurley
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Andrew Jennings
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Lilian Alcaraz
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Dawn Bloomfield
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Michael Briggs
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Stephen Daly
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Terry Panchal
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Vince Russell
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Sharon Wicks
- Charles River Laboratories, 8/9 Spire Green Centre, Flex Meadow, Harlow CM19 5TR, United Kingdom
| | - Harry Finch
- Pulmagen Therapeutics, The Coach House, Grenville Court Britwell Road, Burnham, Slough SL1 8DF, United Kingdom
| | - Mary Fitzgerald
- Pulmagen Therapeutics, The Coach House, Grenville Court Britwell Road, Burnham, Slough SL1 8DF, United Kingdom
| | - Craig Fox
- Pulmagen Therapeutics, The Coach House, Grenville Court Britwell Road, Burnham, Slough SL1 8DF, United Kingdom
| | - Maurizio Delcanale
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| |
Collapse
|
15
|
Michaels WE, Pena-Rasgado C, Kotaria R, Bridges RJ, Hastings ML. Open reading frame correction using splice-switching antisense oligonucleotides for the treatment of cystic fibrosis. Proc Natl Acad Sci U S A 2022; 119:e2114886119. [PMID: 35017302 PMCID: PMC8784102 DOI: 10.1073/pnas.2114886119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022] Open
Abstract
CFTR gene mutations that result in the introduction of premature termination codons (PTCs) are common in cystic fibrosis (CF). This mutation type causes a severe form of the disease, likely because of low CFTR messenger RNA (mRNA) expression as a result of nonsense-mediated mRNA decay, as well as the production of a nonfunctional, truncated CFTR protein. Current therapeutics for CF, which target residual protein function, are less effective in patients with these types of mutations due in part to low CFTR protein levels. Splice-switching antisense oligonucleotides (ASOs), designed to induce skipping of exons in order to restore the mRNA open reading frame, have shown therapeutic promise preclinically and clinically for a number of diseases. We hypothesized that ASO-mediated skipping of CFTR exon 23 would recover CFTR activity associated with terminating mutations in the exon, including CFTR p.W1282X, the fifth most common mutation in CF. Here, we show that CFTR lacking the amino acids encoding exon 23 is partially functional and responsive to corrector and modulator drugs currently in clinical use. ASO-induced exon 23 skipping rescued CFTR expression and chloride current in primary human bronchial epithelial cells isolated from a homozygote CFTR-W1282X patient. These results support the use of ASOs in treating CF patients with CFTR class I mutations in exon 23 that result in unstable CFTR mRNA and truncations of the CFTR protein.
Collapse
Affiliation(s)
- Wren E Michaels
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Science and Medicine, North Chicago, IL 60064
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Science and Medicine, North Chicago, IL 60064
| | - Cecilia Pena-Rasgado
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Science and Medicine, North Chicago, IL 60064
| | - Rusudan Kotaria
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Science and Medicine, North Chicago, IL 60064
| | - Robert J Bridges
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Science and Medicine, North Chicago, IL 60064;
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Science and Medicine, North Chicago, IL 60064;
| |
Collapse
|
16
|
Xia T, Ma J, Sun Y, Sun Y. Androgen receptor suppresses inflammatory response of airway epithelial cells in allergic asthma through MAPK1 and MAPK14. Hum Exp Toxicol 2022; 41:9603271221121320. [PMID: 35982617 DOI: 10.1177/09603271221121320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Dysfunction of airway epithelial cells in patients with asthma is closely with the occurrence and development of allergic asthma. Finding the differences of airway epithelium between asthmatic patients and normal patients is helpful to find out new treatment strategies. METHODS First, three original microarray datasets (GSE89809, GSE41861, GSE104468) from the Gene Expression Omnibus (GEO) dataset were used to assess differentially expressed genes in the epithelial tissues between patients with allergic asthma and healthy controls. Then, 10 ng/mL TGF-β1 treated BEAS-2B cells and rats with ovalbumin induced allergic asthma were performed to confirm our assumption from the gene expression analysis with microarrays. RESULTS Top ten hub significant difference genes were obtained by Cytohubba plug-in from GSE41861, and found that androgen receptor (AR) was closely associated with the mitogen-activated protein kinase (MAPK) pathway, especially MAPK1 and MAPK14. After treated with the TGF-β1 treated BEAS-2B cells and rats with allergic asthma, we found that 5α-dihydrotestosterone (5α-DHT), AR agonist, significantly decreased the Th2 inflammation (IL-25 and IL-33), MAPK1 and MAPK14 proteins expression in vitro and in vivo. The roles of 5α-DHT were similar with the results of chicanine (a p38 MAPK and ERK1/2 inhibitor), but the roles of 5α-DHT were masked by the C16-PAF (a MAPK and MEK/ERK activator) treatment. CONCLUSION Androgen receptor limits the secretion of Th2 inflammatory factors by downregulating MAPK1 and MAPK14 in the TGF-β1 treated BEAS-2B cells and rats with ovalbumin induced allergic asthma, which plays a critical role for the therapeutics of patients with asthma.
Collapse
Affiliation(s)
- T Xia
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, China
| | - J Ma
- Department of Child Healthcare, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, China
| | - Y Sun
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, China
| | - Y Sun
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical College, Yantai, China
| |
Collapse
|
17
|
Zhang HY, Xie QM, Zhao CC, Sha JF, Ruan Y, Wu HM. CpG Oligodeoxynucleotides Attenuate OVA-Induced Allergic Airway Inflammation via Suppressing JNK-Mediated Endoplasmic Reticulum Stress. J Asthma Allergy 2021; 14:1399-1410. [PMID: 34848975 PMCID: PMC8619852 DOI: 10.2147/jaa.s334541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/20/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose CpG-ODN has been found to attenuate allergic airway inflammation in our previous study. Here, we aimed to further investigate whether CpG-ODN exerts such effect via regulating endoplasmic reticulum (ER) stress and revealed the underlying mechanism. Methods Five-week-old C57BL/6 mice were randomly grouped and treated with or without CpG-ODN or/and SP600125. Meantime, RAW264.7 cells were used to investigate the effect of CpG-ODN on OVA-induced ER stress in vitro. The cellularity of bronchoalveolar lavage fluid (BALF) was classified and counted after Wright-Giemsa staining. HE and PAS staining methods were applied to analyze airway inflammation. The protein levels of IL-4, IL-5, IL-13, p-JNK, JNK, CHOP, XBP1, ATF6α and GRP78 in lung tissues were detected by Western blotting. Correspondingly, the ER stress markers were detected by Western blotting and immunofluorescence in RAW264.7 cells. Results In OVA-induced allergic airway inflammation, CpG-ODN significantly suppressed inflammatory cells infiltration, goblet cell hyperplasia and the protein expression of Th2 cytokines. Moreover, OVA exposure strongly increased the activation of ER stress with higher protein expressions of CHOP, XBP1, ATF6α and GRP78. However, these OVA-induced increase of ER stress markers were markedly suppressed by CpG-ODN treatment. In addition, exposure to OVA significantly increased the phosphorylation of JNK, which was significantly reduced by CpG-ODN treatment. Remarkably, single treatment of SP600125, an antagonist of JNK, functioned similarly as CpG-ODN in mitigating allergic airway inflammation and suppressing OVA-induced activation of ER stress; however, no significant synergistic effect was evidenced by combined treatment of SP600125 and CpG-ODN. Furthermore, in OVA-stimulated RAW264.7 cells, we also found that OVA stimulation increased the expressions of ER stress markers, and CpG-ODN significantly reduced their expression levels via suppressing the phosphorylation of JNK. Conclusion These results indicated that CpG-ODN mitigates allergic airway inflammation via suppressing the activation of JNK-medicated ER stress.
Collapse
Affiliation(s)
- Hai-Yun Zhang
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Qiu-Meng Xie
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Cui-Cui Zhao
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Jia-Feng Sha
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Ya Ruan
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Hui-Mei Wu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| |
Collapse
|
18
|
Steele H, Sachen K, McKnight AJ, Soloff R, Herro R. Targeting TL1A/DR3 Signaling Offers a Therapeutic Advantage to Neutralizing IL13/IL4Rα in Muco-Secretory Fibrotic Disorders. Front Immunol 2021; 12:692127. [PMID: 34305924 PMCID: PMC8299868 DOI: 10.3389/fimmu.2021.692127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/17/2021] [Indexed: 01/12/2023] Open
Abstract
Mucus secretion is an important feature of asthma that highly correlates with morbidity. Current therapies, including administration of mucolytics and anti-inflammatory drugs, show limited effectiveness and durability, underscoring the need for novel effective and longer lasting therapeutic approaches. Here we show that mucus production in the lungs is regulated by the TNF superfamily member 15 (TL1A) acting through the mucus-inducing cytokine IL-13. TL1A induces IL13 expression by innate lymphoid cells leading to mucus production, in addition to promoting airway inflammation and fibrosis. Reciprocally, neutralization of IL13 signaling through its receptor (IL4Rα), completely reverses TL1A-induced mucus secretion, while maintaining airway inflammation and fibrosis. Importance of TL1A is further demonstrated using a preclinical asthma model induced by chronic house dust mite exposure where TL1A neutralization by genetic deletion or antagonistic blockade of its receptor DR3 protected against mucus production and fibrosis. Thus, TL1A presents a promising therapeutic target that out benefits IL13 in reversing mucus production, airway inflammation and fibrosis, cardinal features of severe asthma in humans.
Collapse
Affiliation(s)
- Hope Steele
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Kacey Sachen
- Kyowa Kirin Pharmaceutical Research, Inc., La Jolla, CA, United States
| | | | - Rachel Soloff
- Kyowa Kirin Pharmaceutical Research, Inc., La Jolla, CA, United States
| | - Rana Herro
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
19
|
Shin JN, Rao L, Sha Y, Abdel Fattah E, Hyser J, Eissa NT. p38 MAPK Activity Is Required to Prevent Hyperactivation of NLRP3 Inflammasome. THE JOURNAL OF IMMUNOLOGY 2021; 207:661-670. [PMID: 34193605 DOI: 10.4049/jimmunol.2000416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/06/2021] [Indexed: 11/19/2022]
Abstract
Inflammation contributes to the pathogenesis and morbidity of wide spectrum of human diseases. The inflammatory response must be actively controlled to prevent bystander damage to tissues. Yet, the mechanisms controlling excessive inflammatory responses are poorly understood. NLRP3 inflammasome plays an important role in innate immune response to cellular infection or stress. Its activation must be tightly regulated because uncontrolled inflammasome activation is associated with a number of human diseases. p38 MAPK signaling plays an essential role in the regulation of inflammation. The role of p38 MAPK in inflammatory response associated with the expression of proinflammatory molecules is known. However, the anti-inflammatory functions of p38 MAPK are largely unknown. In this study, we show that pharmacologic inhibition or genetic deficiency of p38 MAPK leads to hyperactivation of NLRP3 inflammasome, resulting in enhanced Caspase 1 activation and IL-1β and IL-18 production. The deficiency of p38 MAPK activity induced an increase of cytosolic Ca2+ and excessive mitochondrial Ca2+ uptake, leading to exacerbation of mitochondrial damage, which was associated with hyperactivation of NLRP3 inflammasome. In addition, mice with deficiency of p38 MAPK in granulocytes had evidence of in vivo hyperactivation of NLRP3 inflammasome and were more susceptible to LPS-induced sepsis compared with wild-type mice. Our results suggest that p38 MAPK negatively regulates NLRP3 inflammasome through control of Ca2+ mobilization. Hyperactivity of inflammasome in p38-deficient mice causes lung inflammation and increased susceptibility to septic shock.
Collapse
Affiliation(s)
- Jin Na Shin
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Lang Rao
- Department of Medicine, Baylor College of Medicine, Houston, TX; .,Veterans Administration Long Beach Healthcare System, University of California, Irvine, Irvine, CA.,Southern California Institute for Research and Education, Long Beach, CA; and
| | - Youbao Sha
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | | | - Joseph Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - N Tony Eissa
- Department of Medicine, Baylor College of Medicine, Houston, TX; .,Veterans Administration Long Beach Healthcare System, University of California, Irvine, Irvine, CA
| |
Collapse
|
20
|
MicroRNA Targets for Asthma Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:89-105. [PMID: 33788189 DOI: 10.1007/978-3-030-63046-1_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Asthma is a chronic inflammatory obstructive lung disease that is stratified into endotypes. Th2 high asthma is due to an imbalance of Th1/Th2 signaling leading to abnormally high levels of Th2 cytokines, IL-4, IL-5, and IL-13 and in some cases a reduction in type I interferons. Some asthmatics express Th2 low, Th1/Th17 high phenotypes with or without eosinophilia. Most asthmatics with Th2 high phenotype respond to beta-adrenergic agonists, muscarinic antagonists, and inhaled corticosteroids. However, 5-10% of asthmatics are not well controlled by these therapies despite significant advances in lung immunology and the pathogenesis of severe asthma. This problem is being addressed by developing novel classes of anti-inflammatory agents. Numerous studies have established efficacy of targeting pro-inflammatory microRNAs in mouse models of mild/moderate and severe asthma. Current approaches employ microRNA mimics and antagonists designed for use in vivo. Chemically modified oligonucleotides have enhanced stability in blood, increased cell permeability, and optimized target specificity. Delivery to lung tissue limits clinical applications, but it is a tractable problem. Future studies need to define the most effective microRNA targets and effective delivery systems. Successful oligonucleotide drug candidates must have adequate lung cell uptake, high target specificity, and efficacy with tolerable off-target effects.
Collapse
|
21
|
Pelaia C, Vatrella A, Gallelli L, Lombardo N, Sciacqua A, Savino R, Pelaia G. Role of p38 Mitogen-Activated Protein Kinase in Asthma and COPD: Pathogenic Aspects and Potential Targeted Therapies. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:1275-1284. [PMID: 33790539 PMCID: PMC8001041 DOI: 10.2147/dddt.s300988] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022]
Abstract
Among the various members of the mitogen-activated protein kinase (MAPK) family, p38 MAPK subgroup is the most involved in airway and lung inflammation underlying asthma and chronic obstructive pulmonary disease (COPD). In particular, several environmental agents including aeroallergens, cigarette smoke, airborne pollutants, viral and bacterial pathogens activate the p38α isoform which in turn up-regulates the expression of multiple proinflammatory cytokines and chemokines, as well as the production of some fibrogenic factors. Therefore, p38 MAPK-induced bronchial inflammation and remodelling significantly contribute to the development, persistence and amplification of airflow limitation, which is the hallmark of asthma and COPD. Such advances in our understanding of p38 role in the pathobiology of the above widespread, chronic obstructive respiratory diseases, have led to consider p38 MAPK as a suitable molecular target for novel treatment strategies. Indeed, many studies have been carried out in both animal and clinical settings, with the aim of evaluating the potential therapeutic effects of p38 MAPK inhibitors in both asthma and COPD.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Luca Gallelli
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Nicola Lombardo
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Rocco Savino
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
22
|
Hirai K, Shirai T, Shimoshikiryo T, Ueda M, Gon Y, Maruoka S, Itoh K. Circulating microRNA-15b-5p as a biomarker for asthma-COPD overlap. Allergy 2021; 76:766-774. [PMID: 32713026 DOI: 10.1111/all.14520] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/24/2020] [Accepted: 07/09/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND It remains unclear how to characterize different subtypes of asthma and chronic obstructive pulmonary disease (COPD). We previously described serum periostin and chitinase-3-like protein 1 (YKL-40) as useful markers for asthma-COPD overlap (ACO). MicroRNAs (miRNAs) are now recognized as markers for identifying the pathophysiological features in several diseases. This study aimed to identify circulating miRNAs that could discriminate patients with ACO from patients with asthma or COPD. METHODS This study included two independent cohorts. First, we screened 84 miRNAs for expression levels in patients with ACO (n = 6) or asthma (n = 6) using a quantitative real-time PCR array. The miRNAs showing at least a 2-fold difference in the discovery phase were analyzed in 30 patients each with asthma, COPD, or ACO in the replication phase. The diagnostic accuracy was evaluated using the area under the receiver operating characteristic curve (AUROC). RESULTS Nine miRNAs were identified in the discovery phase. Five of these miRNAs (miR-148a-3p, miR-15b-5p, miR-223-3p, miR-23a-3p, and miR-26b-5p) had lower levels in ACO patients and could discriminate between ACO patients and patients with either asthma or COPD. miR-15b-5p was the most accurate miRNA for the discrimination of patients with ACO (AUROC, 0.71). Moreover, the combined assessment of miR-15b-5p, serum periostin, and YKL-40 (AUROC, 0.80) improved diagnostic accuracy for ACO compared with the combined model of periostin and YKL-40 (AUROC, 0.69). CONCLUSIONS Circulating miR-15b-5p is a potential marker for identifying patients with ACO. By elucidating the molecular pathways controlled by miRNAs, we may better understand the pathophysiology of ACO.
Collapse
Affiliation(s)
- Keita Hirai
- Department of Clinical Pharmacology & Genetics School of Pharmaceutical Sciences University of Shizuoka Shizuoka Japan
- Laboratory of Clinical Pharmacogenomics Shizuoka General Hospital Shizuoka Japan
| | - Toshihiro Shirai
- Department of Respiratory Medicine Shizuoka General Hospital Shizuoka Japan
| | - Takayuki Shimoshikiryo
- Department of Clinical Pharmacology & Genetics School of Pharmaceutical Sciences University of Shizuoka Shizuoka Japan
| | - Megumi Ueda
- Department of Clinical Pharmacology & Genetics School of Pharmaceutical Sciences University of Shizuoka Shizuoka Japan
| | - Yasuhiro Gon
- Division of Respiratory Medicine Department of Internal Medicine Nihon University School of Medicine Tokyo Japan
| | - Shuichiro Maruoka
- Division of Respiratory Medicine Department of Internal Medicine Nihon University School of Medicine Tokyo Japan
| | - Kunihiko Itoh
- Department of Clinical Pharmacology & Genetics School of Pharmaceutical Sciences University of Shizuoka Shizuoka Japan
- Laboratory of Clinical Pharmacogenomics Shizuoka General Hospital Shizuoka Japan
| |
Collapse
|
23
|
Xu Y, Thakur A, Zhang Y, Foged C. Inhaled RNA Therapeutics for Obstructive Airway Diseases: Recent Advances and Future Prospects. Pharmaceutics 2021; 13:pharmaceutics13020177. [PMID: 33525500 PMCID: PMC7912103 DOI: 10.3390/pharmaceutics13020177] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/20/2021] [Accepted: 01/23/2021] [Indexed: 02/06/2023] Open
Abstract
Obstructive airway diseases, e.g., chronic obstructive pulmonary disease (COPD) and asthma, represent leading causes of morbidity and mortality worldwide. However, the efficacy of currently available inhaled therapeutics is not sufficient for arresting disease progression and decreasing mortality, hence providing an urgent need for development of novel therapeutics. Local delivery to the airways via inhalation is promising for novel drugs, because it allows for delivery directly to the target site of action and minimizes systemic drug exposure. In addition, novel drug modalities like RNA therapeutics provide entirely new opportunities for highly specific treatment of airway diseases. Here, we review state of the art of conventional inhaled drugs used for the treatment of COPD and asthma with focus on quality attributes of inhaled medicines, and we outline the therapeutic potential and safety of novel drugs. Subsequently, we present recent advances in manufacturing of thermostable solid dosage forms for pulmonary administration, important quality attributes of inhalable dry powder formulations, and obstacles for the translation of inhalable solid dosage forms to the clinic. Delivery challenges for inhaled RNA therapeutics and delivery technologies used to overcome them are also discussed. Finally, we present future prospects of novel inhaled RNA-based therapeutics for treatment of obstructive airways diseases, and highlight major knowledge gaps, which require further investigation to advance RNA-based medicine towards the bedside.
Collapse
Affiliation(s)
- You Xu
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (Y.X.); (A.T.); (Y.Z.)
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (Y.X.); (A.T.); (Y.Z.)
| | - Yibang Zhang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (Y.X.); (A.T.); (Y.Z.)
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (Y.X.); (A.T.); (Y.Z.)
- Correspondence: ; Tel.: +45-3533-6402
| |
Collapse
|
24
|
Michaels WE, Bridges RJ, Hastings ML. Antisense oligonucleotide-mediated correction of CFTR splicing improves chloride secretion in cystic fibrosis patient-derived bronchial epithelial cells. Nucleic Acids Res 2020; 48:7454-7467. [PMID: 32520327 PMCID: PMC7367209 DOI: 10.1093/nar/gkaa490] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 05/22/2020] [Accepted: 06/05/2020] [Indexed: 12/27/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, encoding an anion channel that conducts chloride and bicarbonate across epithelial membranes. Mutations that disrupt pre-mRNA splicing occur in >15% of CF cases. One common CFTR splicing mutation is CFTR c.3718-2477C>T (3849+10 kb C>T), which creates a new 5′ splice site, resulting in splicing to a cryptic exon with a premature termination codon. Splice-switching antisense oligonucleotides (ASOs) have emerged as an effective therapeutic strategy to block aberrant splicing. We test an ASO targeting the CFTR c.3718-2477C>T mutation and show that it effectively blocks aberrant splicing in primary bronchial epithelial (hBE) cells from CF patients with the mutation. ASO treatment results in long-term improvement in CFTR activity in hBE cells, as demonstrated by a recovery of chloride secretion and apical membrane conductance. We also show that the ASO is more effective at recovering chloride secretion in our assay than ivacaftor, the potentiator treatment currently available to these patients. Our findings demonstrate the utility of ASOs in correcting CFTR expression and channel activity in a manner expected to be therapeutic in patients.
Collapse
Affiliation(s)
- Wren E Michaels
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Robert J Bridges
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| |
Collapse
|
25
|
Keenan MM, Huang L, Jordan NJ, Wong E, Cheng Y, Valley HC, Mahiou J, Liang F, Bihler H, Mense M, Guo S, Monia BP. Nonsense-mediated RNA Decay Pathway Inhibition Restores Expression and Function of W1282X CFTR. Am J Respir Cell Mol Biol 2020; 61:290-300. [PMID: 30836009 DOI: 10.1165/rcmb.2018-0316oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The recessive genetic disease cystic fibrosis (CF) is caused by loss-of-function mutations in the CFTR (CF transmembrane conductance regulator) gene. Approximately 10% of patients with CF have at least one allele with a nonsense mutation in CFTR. Nonsense mutations generate premature termination codons that can subject mRNA transcripts to rapid degradation through the nonsense-mediated mRNA decay (NMD) pathway. Currently, there are no approved therapies that specifically target nonsense mutations in CFTR. Here, we identified antisense oligonucleotides (ASOs) that target the NMD factor SMG1 to inhibit the NMD pathway, and determined their effects on the W1282X CFTR mutation. First, we developed and validated two in vitro models of the W1282X CFTR mutation. Next, we treated these cells with antisense oligonucleotides to inhibit NMD and measured the effects of these treatments on W1282X expression and function. SMG1-ASO-mediated NMD inhibition upregulated the RNA, protein, and surface-localized protein expression of the truncated W1282X gene product. Additionally, these ASOs increased the CFTR chloride channel function in cells homozygous for the W1282X mutation. Our approach suggests a new therapeutic strategy for patients harboring nonsense mutations and may be beneficial as a single agent in patients with CF and the W1282X mutation.
Collapse
Affiliation(s)
| | - Lulu Huang
- Ionis Pharmaceuticals, Carlsbad, California; and
| | - Nikole J Jordan
- Cystic Fibrosis Foundation Therapeutics Lab, Cystic Fibrosis Foundation, Lexington, Massachusetts
| | - Eric Wong
- Cystic Fibrosis Foundation Therapeutics Lab, Cystic Fibrosis Foundation, Lexington, Massachusetts
| | - Yi Cheng
- Cystic Fibrosis Foundation Therapeutics Lab, Cystic Fibrosis Foundation, Lexington, Massachusetts
| | - Hillary C Valley
- Cystic Fibrosis Foundation Therapeutics Lab, Cystic Fibrosis Foundation, Lexington, Massachusetts
| | - Jerome Mahiou
- Cystic Fibrosis Foundation Therapeutics Lab, Cystic Fibrosis Foundation, Lexington, Massachusetts
| | - Feng Liang
- Cystic Fibrosis Foundation Therapeutics Lab, Cystic Fibrosis Foundation, Lexington, Massachusetts
| | - Hermann Bihler
- Cystic Fibrosis Foundation Therapeutics Lab, Cystic Fibrosis Foundation, Lexington, Massachusetts
| | - Martin Mense
- Cystic Fibrosis Foundation Therapeutics Lab, Cystic Fibrosis Foundation, Lexington, Massachusetts
| | - Shuling Guo
- Ionis Pharmaceuticals, Carlsbad, California; and
| | | |
Collapse
|
26
|
Ramelli SC, Comer BS, McLendon JM, Sandy LL, Ferretti AP, Barrington R, Sparks J, Matar M, Fewell J, Gerthoffer WT. Nanoparticle Delivery of Anti-inflammatory LNA Oligonucleotides Prevents Airway Inflammation in a HDM Model of Asthma. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1000-1014. [PMID: 32044723 PMCID: PMC7013130 DOI: 10.1016/j.omtn.2019.12.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/16/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023]
Abstract
To address the problem of poor asthma control due to drug resistance, an antisense oligonucleotide complementary to mmu-miR-145a-5p (antimiR-145) was tested in a house dust mite mouse model of mild/moderate asthma. miR-145 was targeted to reduce inflammation, regulate epithelial-mesenchymal transitions, and promote differentiation of structural cells. In addition, several chemical variations of a nontargeting oligonucleotide were tested to define sequence-dependent effects of the miRNA antagonist. After intravenous administration, oligonucleotides complexed with a pegylated cationic lipid nanoparticle distributed to most cells in the lung parenchyma but were not present in smooth muscle or the mucosal epithelium of the upper airways. Treatment with antimiR-145 and a nontargeting oligonucleotide both reduced eosinophilia, reduced obstructive airway remodeling, reduced mucosal metaplasia, and reduced CD68 immunoreactivity. Poly(A) RNA-seq verified that antimiR-145 increased levels of many miR-145 target transcripts. Genes upregulated in human asthma and the mouse model of asthma were downregulated by oligonucleotide treatments. However, both oligonucleotides significantly upregulated many genes of interferon signaling pathways. These results establish effective lung delivery and efficacy of locked nucleic acid/DNA oligonucleotides administered intravenously, and suggest that some of the beneficial effects of oligonucleotide therapy of lung inflammation may be due to normalization of interferon response pathways.
Collapse
Affiliation(s)
- Sabrina C Ramelli
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Brian S Comer
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Jared M McLendon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Lydia L Sandy
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Andrew P Ferretti
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, USA
| | - Robert Barrington
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, USA
| | - Jeff Sparks
- Celsion Corporation, 601 Genome Way, Huntsville, AL, USA
| | - Majed Matar
- Celsion Corporation, 601 Genome Way, Huntsville, AL, USA
| | - Jason Fewell
- Celsion Corporation, 601 Genome Way, Huntsville, AL, USA
| | - William T Gerthoffer
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA; Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, USA.
| |
Collapse
|
27
|
Pelaia C, Vatrella A, Crimi C, Gallelli L, Terracciano R, Pelaia G. Clinical relevance of understanding mitogen-activated protein kinases involved in asthma. Expert Rev Respir Med 2020; 14:501-510. [PMID: 32098546 DOI: 10.1080/17476348.2020.1735365] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Mitogen-activated protein kinases (MAPKs) are a large family of evolutionary conserved intracellular enzymes that play a pivotal role in signaling pathways mediating the biologic actions of a wide array of extracellular stimuli.Areas covered: MAPKs are implicated in most pathogenic events involved in asthma, including both inflammatory and structural changes occurring in the airways. Indeed, MAPKs are located at the level of crucial convergence points within the signal transduction networks activated by many cytokines, chemokines, growth factors, and other inducers of bronchial inflammation and remodeling such as immunoglobulin E (IgE) and oxidative stress.Expert opinion: Therefore, given the growing importance of MAPKs in asthma pathobiology, these signaling enzymes are emerging as key intracellular pathways whose upstream activation can be inhibited by biological drugs such as anti-cytokines and anti-IgE.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Claudia Crimi
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Luca Gallelli
- Department of Health Science, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Rosa Terracciano
- Department of Health Science, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Girolamo Pelaia
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
28
|
Duan J, Xie J, Deng T, Xie X, Liu H, Li B, Chen M. Exposure to both formaldehyde and high relative humidity exacerbates allergic asthma by activating the TRPV4-p38 MAPK pathway in Balb/c mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 256:113375. [PMID: 31662264 DOI: 10.1016/j.envpol.2019.113375] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/20/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
Some studies have indicated that formaldehyde, a ubiquitous environmental pollutant, can induce or aggravate allergic asthma. Epidemiological studies have also shown that the relative humidity indoors may be an independent and a key factor associated with the aggravation of allergic asthma. However, the synergy of humidity and formaldehyde on allergic asthma and the mechanism underlying this effect remain largely unknown. In this study, we aim to determine the effect of high relative humidity and/or formaldehyde exposure on allergic asthma and explore the underlying mechanisms. Male Balb/c mice were modeled with ovalbumin (OVA) and exposure to 0.5 mg/m3 formaldehyde and/or different relative humidity (60%/75%/90%). Histopathological changes, pulmonary function, Th1/Th2 balance, the status of mucus hypersecretion and the levels of inflammatory factors were detected to assess the exacerbation of allergic asthma. The levels of the transient receptor potential vanilloid 4 (TRPV4), calcium ion and the activation of p38 mitogen-activated protein kinases (p38 MAPK) were detected to explore the underlying mechanisms. The results showed that exposure to high relative humidity or to 0.5 mg/m3 formaldehyde alone had a slight, but not significant, affect on allergic asthma. However, the pathological response and airway hyperresponsiveness (AHR) were greatly aggravated by simultaneous exposure to 0.5 mg/m3 formaldehyde and 90% relative humidity. Blocking TRPV4or p38 MAPK using HC-067047 and SB203580 respectively, effectively alleviated the exacerbation of allergic asthma induced by this simultaneous exposure to formaldehyde and high relative humidity. The results show that when formaldehyde and high relative humidity are present this can enhance the activation of the TRPV4 ion channel in the lung leading to the aggravation of the p38 MAPK activation, resulting in the exacerbation of inflammation and hypersecretion of mucus in the airways.
Collapse
Affiliation(s)
- Jiufei Duan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Jing Xie
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Ting Deng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Xiaoman Xie
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Hong Liu
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Chongqing 400045, China
| | - Baizhan Li
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Chongqing 400045, China
| | - Mingqing Chen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China.
| |
Collapse
|
29
|
Li J, Zheng M, Wang C, Jiang J, Xu C, Li L, Li L, Yan G, Jin Y. Cryptotanshinone attenuates allergic airway inflammation through negative regulation of NF-κB and p38 MAPK. Biosci Biotechnol Biochem 2019; 84:268-278. [PMID: 31690224 DOI: 10.1080/09168451.2019.1687280] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This study is to determine the role and mechanism of cryptotanshinone (CTS) in allergic airway inflammation. Asthma induced by OVA was established in BALB/c mice. We found increased airway hyperresponsiveness (AHR), increased inflammatory cell infiltration, elevated levels of TNF-α, interleukin-1β (IL-1β), IL-4, IL-5, IL-6 and IL-13, decreased interferon gamma (IFN-γ) in lung tissue, increased content of total immunoglobulin E (IgE), OVA specific IgE, Eotaxin, ICAM-1, VCAM-1, nuclear factor-kappaB (NF-κB) and phosphorylation of p38 MAPK in lung tissue. However, the administration of CTS significantly decreased AHR in asthmatic mice, reduced inflammation around the bronchioles and inflammatory cells around airway, regulated cytokine production, reduced the total IgE and OVA-specific IgE levels, and inhibited NF-κB activation and p38 MAPK phosphorylation. In vitro experiments in 16 HBE cells revealed that CTS attenuated CAM-1 and IL-6 expression. These results indicate that CTS alleviates allergic airway inflammation by modulating p38 MAPK phosphorylation and NF-κB activation.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Mingyu Zheng
- College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Chongyang Wang
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Jingzhi Jiang
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Chang Xu
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Li Li
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Liangchang Li
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Guanghai Yan
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, P.R. China
| | - Yongde Jin
- Department of Otolaryngology-Head and Neck Surgery, Yanbian University Hospital, Yanji, P.R. China
| |
Collapse
|
30
|
Strâmbu IR, Kobalava ZD, Magnusson BP, MacKinnon A, Parkin JM. Phase II Study of Single/Repeated Doses of Acumapimod (BCT197) to Treat Acute Exacerbations of COPD. COPD 2019; 16:344-353. [PMID: 31682162 DOI: 10.1080/15412555.2019.1682535] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mitogen-activated protein kinase p38 is a key regulator in the inflammation pathway and is activated in the lungs of chronic obstructive pulmonary disease (COPD) patients. Acumapimod is a potent, selective, oral, p38 inhibitor under investigation for treatment of acute exacerbations of COPD (AECOPD). In this Phase II, double-blind, randomized, placebo-controlled dose-exploration study of acumapimod in patients with moderate or severe AECOPD (NCT01332097), patients presenting with AECOPD were randomized to receive single-dose acumapimod (20 mg or 75 mg) on Day 1, repeated single-dose acumapimod (20 mg or 75 mg) on Days 1 and 6, oral prednisone 40 mg (10 days), or placebo. Primary outcome: improvement in forced expiratory volume in 1 s (FEV1) versus placebo at Day 5 (single doses) and Day 10 (repeated doses). N = 183 patients were randomized; 169 (92%) patients completed the study. Although the primary endpoint (FEV1 at Day 10) was not met (p = 0.082), there was a significant improvement in FEV1 with acumapimod repeat-dose 75 mg versus placebo at Day 8 (p = 0.022) which, though not a prespecified endpoint, was part of an overall trend. Differences at lower doses did not achieve significance. Mean change in FEV1 AUC from baseline to Day 14 in the 75 mg repeat-dose group was significantly higher versus placebo (p = 0.02), prednisone (p = 0.01), and 20 mg single-dose groups (p = 0.015) (post-hoc analysis). EXACT-PRO showed numerical differences versus placebo that did not reach significance. Acumapimod was well tolerated. In conclusion, repeated single-dose acumapimod showed a clinically relevant improvement in FEV1 over placebo at Day 8, along with consistent numerical differences in EXACT-PRO. These data can be used to determine dose regimens for a proof-of-clinical-concept trial.
Collapse
Affiliation(s)
- Irina R Strâmbu
- National Institute of Pneumology "Marius Nasta", Bucharest, Romania
| | | | | | | | | |
Collapse
|
31
|
Zhang Y, Li X, He M, Zhang G, Bao W, Fei X, Zhang X, Zhang M, Zhang P. The effects of neutralizing anti-murine interleukin-17A monoclonal antibody on ozone-induced inflammation and glucocorticoids insensitivity in a murine model of asthma. Biomed Pharmacother 2019; 114:108786. [PMID: 30917335 DOI: 10.1016/j.biopha.2019.108786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 03/09/2019] [Accepted: 03/13/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Exposure to ozone contributed to the worsening of inflammation and glucocorticoids insensitivity in OVA-challenged asthma. Interleukin-17A participates centrally in stages of the inflammatory response and glucocorticoids insensitivity. In this study, the effect of neutralizing anti-murine interleukin-17A monoclonal antibody (IL-17A mAb) on inflammation and glucocorticoids insensitivity in ozone-exposed and ovalbumin (OVA)-challenged mice was investigated. METHODS Mice were sensitized and challenged with OVA and then exposed to ozone. Dexamethasone (Dex) and IL-17A mAb were administrated in corresponding periods. RESULTS Compared with OVA-challenged mice, combination administration of ozone exposure and OVA challenge increased the recruitment of inflammatory cells in bronchoalveolar lavage fluid, enhanced the inflammation scores and levels of inflammatory cytokines and IL-17A mRNA, and caused the activation of p38 MAPK together with down regulation of glucocorticoids recepters (GR) in lung tissue. Monotherapy of IL-17A mAb partially attenuated lung inflammation in OVA-challenged and ozone-exposed mice, while the combination treatment of Dex and IL-17A mAb effectively reduced lung inflammation, inactivated p38 MAPK and up regulated GR in lung tissue. CONCLUSIONS Ozone exposure worsened OVA-challenged airway inflammation, activation of p38 MAPK and down regulation of GR in OVA-sensitized and -challenged mice, which was effectively counteracted by IL-17A mAb, and combination treatment of IL-17A mAb and Dex shows profound efficacy in inhibiting airway inflammation and improving glucocorticoids insensitivity synergistically.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xingjing Li
- Department of Respiratory Medicine, The Shanghai First People's Hospital of Baoshan Branch, Shanghai, China
| | - Mudan He
- Department of Respiratory Medicine, The Shanghai First People's Hospital of Baoshan Branch, Shanghai, China
| | - Guoqing Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wuping Bao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Fei
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xue Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Pengyu Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
32
|
Interleukin-35 expression protects against cigarette smoke-induced lung inflammation in mice. Biomed Pharmacother 2018; 110:727-732. [PMID: 30554110 DOI: 10.1016/j.biopha.2018.12.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 11/24/2022] Open
Abstract
Cigarette smoke (CS) is a very important cause of pulmonary inflammatory diseases. Interleukin (IL)-35 is a novel anti-inflammatory cytokine but its role in CS-mediated lung inflammation remains unclear. In the present study, we examined the effect of IL-35 expression on CS-induced lung inflammation in mice. A plasmid DNA expressing IL-35 was injected into mice via a hydrodynamic-based gene delivery that were subsequently exposed to CS three times a day for 5 days. We found that IL-35 expression inhibited pulmonary inflammatory infiltration, lung tissue lesions, mucus secretion, and myeloperoxidase activity in CS-treated mice. Moreover, IL-35 expression decreased the production of IL-1β, tumor necrosis factor-α, IL-6, and IL-17, but increased the level of IL-10 in bronchoalveolar lavage fluids and lung tissues from CS-challenged mice. These results suggest that in vivo expression of IL-35 can protect against CS-induced lung inflammation and may be a therapeutic target in CS-related pulmonary diseases.
Collapse
|
33
|
Piao H, Choi YH, Li H, Wang C, Xian Z, Ogasawara M, Jiang J, Li L, Yamauchi K, Yan G. Recombinant pyrin domain protein attenuates allergic inflammation by suppressing NF-κB pathway in asthmatic mice. Scand J Immunol 2018; 89:e12720. [PMID: 30589094 DOI: 10.1111/sji.12720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/26/2018] [Accepted: 09/29/2018] [Indexed: 01/09/2023]
Abstract
Pyrin domain (PYD), a subclass of protein motif known as the death fold, is frequently involved in inflammation and immune responses. PYD modulates nuclear factor-kappa B (NF-κB) signalling pathway upon various stimuli. Herein, a novel recombinant pyrin domain protein (RPYD) was generated. Its role and mechanism in inflammatory response in an ovalbumin (OVA) induced asthma model was investigated. After OVA challenge, there was inflammatory cell infiltration in the lung, as well as airway hyper-responsiveness (AHR) to inhaled methacholine. In addition, eosinophils increased in the bronchoalveolar lavage fluids, alone with the elevated levels of Th-2 type cytokines [interleukin (IL)-4, IL-5 and IL-13], eotaxin, and adhesion molecules. However, the transnasal administration of RPYD before the OVA challenge significantly inhibited these asthmatic reactions. Moreover, RPYD markedly suppressed NF-κB translocation, reduced phosphorylation of p38 MAPK, and thus attenuated the expression of intercellular adhesion molecule 1 and IL-6 in the BEAS-2B cells stimulated by proinflammatory cytokines in vitro. These findings indicate that RPYD can protect asthma host from OVA-induced airway inflammation and AHR via down-regulation of NF-κB and p38 MAPK activities. RPYD may be used as a potential medicine for the treatment of asthma in clinic.
Collapse
Affiliation(s)
- Hongmei Piao
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, YanJi, Jilin, China
| | - Yun Ho Choi
- Department of Anatomy, Medical School, Institute for Medical Sciences, Chonbuk National University, Jeonju, Jeonbuk, Korea
| | - Hongmei Li
- Administration of Traditional Chinese Medicine of JiLin Province, Changchun, China
| | - Chongyang Wang
- Department of Anatomy and Histology and Embryology, Yanbian University Medical College, YanJi, Jilin, China
| | - Zhemin Xian
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, YanJi, Jilin, China
| | - Masahito Ogasawara
- Division of Pharmacology, Department of Integrated Life Science, Ehime University School of Medicine, Ehime, Japan
| | - Jingzhi Jiang
- Department of Anatomy and Histology and Embryology, Yanbian University Medical College, YanJi, Jilin, China
| | - Liangchang Li
- Department of Anatomy and Histology and Embryology, Yanbian University Medical College, YanJi, Jilin, China
| | - Kohei Yamauchi
- Division of Pulmonary Medicine, Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Guanghai Yan
- Department of Anatomy and Histology and Embryology, Yanbian University Medical College, YanJi, Jilin, China
| |
Collapse
|
34
|
Martinovich KM, Shaw NC, Kicic A, Schultz A, Fletcher S, Wilton SD, Stick SM. The potential of antisense oligonucleotide therapies for inherited childhood lung diseases. Mol Cell Pediatr 2018; 5:3. [PMID: 29411170 PMCID: PMC5801198 DOI: 10.1186/s40348-018-0081-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/25/2018] [Indexed: 01/05/2023] Open
Abstract
Antisense oligonucleotides are an emerging therapeutic option to treat diseases with known genetic origin. In the age of personalised medicines, antisense oligonucleotides can sometimes be designed to target and bypass or overcome a patient's genetic mutation, in particular those lesions that compromise normal pre-mRNA processing. Antisense oligonucleotides can alter gene expression through a variety of mechanisms as determined by the chemistry and antisense oligomer design. Through targeting the pre-mRNA, antisense oligonucleotides can alter splicing and induce a specific spliceoform or disrupt the reading frame, target an RNA transcript for degradation through RNaseH activation, block ribosome initiation of protein translation or disrupt miRNA function. The recent accelerated approval of eteplirsen (renamed Exondys 51™) by the Food and Drug Administration, for the treatment of Duchenne muscular dystrophy, and nusinersen, for the treatment of spinal muscular atrophy, herald a new and exciting era in splice-switching antisense oligonucleotide applications to treat inherited diseases. This review considers the potential of antisense oligonucleotides to treat inherited lung diseases of childhood with a focus on cystic fibrosis and disorders of surfactant protein metabolism.
Collapse
Affiliation(s)
- Kelly M. Martinovich
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Centre for Comparative Genomics, Murdoch University, Murdoch, Western Australia 6150 Australia
| | - Nicole C. Shaw
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Centre for Comparative Genomics, Murdoch University, Murdoch, Western Australia 6150 Australia
| | - Anthony Kicic
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Subiaco, Western Australia 6008 Australia
- School of Public Health, Curtin University, Bentley, Western Australia 6102 Australia
| | - André Schultz
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Subiaco, Western Australia 6008 Australia
| | - Sue Fletcher
- Perron Institute for Neurological and Translational Sciences, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Centre for Comparative Genomics, Murdoch University, Murdoch, Western Australia 6150 Australia
| | - Steve D. Wilton
- Perron Institute for Neurological and Translational Sciences, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Centre for Comparative Genomics, Murdoch University, Murdoch, Western Australia 6150 Australia
| | - Stephen M. Stick
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia 6009 Australia
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Subiaco, Western Australia 6008 Australia
| |
Collapse
|
35
|
Uemura Y, Hagiwara K, Kobayashi K. The intratracheal administration of locked nucleic acid containing antisense oligonucleotides induced gene silencing and an immune-stimulatory effect in the murine lung. PLoS One 2017; 12:e0187286. [PMID: 29107995 PMCID: PMC5673232 DOI: 10.1371/journal.pone.0187286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 10/17/2017] [Indexed: 12/21/2022] Open
Abstract
Locked nucleic acid containing antisense oligonucleotides (LNA-ASOs) have the potential to modulate the disease-related gene expression by the RNaseH-dependent degradation of mRNAs. Pulmonary drug delivery has been widely used for the treatment of lung disease. Thus, the inhalation of LNA-ASOs is expected to be an efficient therapy that can be applied to several types of lung disease. Because the lung has a distinct immune system against pathogens, the immune-stimulatory effect of LNA-ASOs should be considered for the development of novel inhaled LNA-ASOs therapies. However, there have been no reports on the relationship between knock-down (KD) and the immune-stimulatory effects of inhaled LNA-ASOs in the lung. In this report, LNA-ASOs targeting Scarb1 (Scarb1-ASOs) or negative control LNA-ASOs targeting ApoB (ApoB-ASOs) were intratracheally administered to mice to investigate the KD of the gene expression and the immune-stimulatory effects in the lung. We confirmed that the intratracheal administration of Scarb1-ASOs exerted a KD effect in the lung without a drug delivery system. On the other hand, both Scarb1-ASOs and ApoB-ASOs induced neutrophilic infiltration in the alveoli and increased the expression levels of G-CSF and CXCL1 in the lung. The dose required for KD was the same as the dose that induced the neutrophilic immune response. In addition, in our in vitro experiments, Scarb1-ASOs did not increase the G-CSF or CXCL1 expression in primary lung cells, even though Scarb1-ASOs exerted a strong KD effect. Hence, we hypothesize that inhaled LNA-ASOs have the potential to exert a KD effect in the lung, but that they may be associated with a risk of immune stimulation. Further studies about the mechanism underlying the immune-stimulatory effect of LNA-ASOs is necessary for the development of novel inhaled LNA-ASO therapies.
Collapse
Affiliation(s)
- Yasunori Uemura
- Immunology & Allergy R&D Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Shizuoka, Japan
- * E-mail:
| | - Kenji Hagiwara
- Innovative Technology Labs, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Machida, Tokyo, Japan
| | - Katsuya Kobayashi
- Immunology & Allergy R&D Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Shizuoka, Japan
| |
Collapse
|
36
|
Pazhoohan S, Raoufy MR, Javan M, Hajizadeh S. Effect of Rho-kinase inhibition on complexity of breathing pattern in a guinea pig model of asthma. PLoS One 2017; 12:e0187249. [PMID: 29088265 PMCID: PMC5663484 DOI: 10.1371/journal.pone.0187249] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022] Open
Abstract
Asthma represents an episodic and fluctuating behavior characterized with decreased complexity of respiratory dynamics. Several evidence indicate that asthma severity or control is associated with alteration in variability of lung function. The pathophysiological basis of alteration in complexity of breathing pattern in asthma has remained poorly understood. Regarding the point that Rho-kinase is involved in pathophysiology of asthma, in present study we investigated the effect of Rho-kinase inhibition on complexity of respiratory dynamics in a guinea pig model of asthma. Male Dunkin Hartley guinea pigs were exposed to 12 series of inhalations with ovalbumin or saline. Animals were treated by the Rho-kinase inhibitor Y-27632 (1mM aerosols) prior to each allergen challenge. We recorded respiration of conscious animals using whole-body plethysmography. Exposure to ovalbumin induced lung inflammation, airway hyperresponsiveness and remodeling including goblet cell hyperplasia, increase in the thickness of airways smooth muscles and subepithelial collagen deposition. Complexity analysis of respiratory dynamics revealed a dramatic decrease in irregularity of respiratory rhythm representing less complexity in asthmatic guinea pigs. Inhibition of Rho-kinase reduced the airway remodeling and hyperreponsiveness, but had no significant effect on lung inflammation and complexity of respiratory dynamics in asthmatic animals. It seems that airway hyperresponsiveness and remodeling do not significantly affect the complexity of respiratory dynamics. Our results suggest that inflammation might be the probable cause of shift in the respiratory dynamics away from the normal fluctuation in asthma.
Collapse
Affiliation(s)
- Saeed Pazhoohan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- * E-mail: (MRR); (SH)
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sohrab Hajizadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- * E-mail: (MRR); (SH)
| |
Collapse
|
37
|
Khorasanizadeh M, Eskian M, Gelfand EW, Rezaei N. Mitogen-activated protein kinases as therapeutic targets for asthma. Pharmacol Ther 2017; 174:112-126. [DOI: 10.1016/j.pharmthera.2017.02.024] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
38
|
Wijerathne CUB, Seo CS, Song JW, Park HS, Moon OS, Won YS, Kwon HJ, Son HY. Isoimperatorin attenuates airway inflammation and mucus hypersecretion in an ovalbumin-induced murine model of asthma. Int Immunopharmacol 2017; 49:67-76. [PMID: 28551494 DOI: 10.1016/j.intimp.2017.05.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/26/2017] [Accepted: 05/10/2017] [Indexed: 01/22/2023]
Abstract
Isoimperatorin (IMP), an active natural furocoumarin, has numerous pharmacologic effects, including anti-inflammatory, analgesic, antispasmodic, and anticancer activities. This study aimed to evaluate the preventive activity of IMP in an ovalbumin (OVA)-induced murine model of asthma and to investigate its possible molecular mechanisms. Female BALB/c mice were sensitized on days 0 and 14 via intraperitoneal injection of 20μg OVA. On days 21-23 after the initial sensitization, the mice received an airway challenge with OVA (1% w/v in PBS) for 1h; meanwhile, IMP (10 or 30mg/kg once daily) was administered by gavage on days 18-23. Our results revealed that IMP significantly lowered the productions of interleukin (IL)-4, IL-5, IL-13, eotaxin, and immunoglobulin (Ig)E in bronchoalveolar lavage fluid (BALF), plasma, or lung tissues. Histological studies showed that IMP inhibited OVA-induced inflammatory cell infiltration and mucus production in the respiratory tract. In addition, pretreatment with IMP suppressed the activation of p38 mitogen-activated protein kinase (p38 MAPK), extracellular-signal-regulated kinases 1/2 (ERK1/2), and nuclear factor-κB (NF-κB). Together, these results suggest that IMP effectively inhibits airway inflammation and mucus hypersecretion by downregulating the levels of Th2 cytokines and inhibiting NF-κB and MAPK pathways.
Collapse
Affiliation(s)
- Charith U B Wijerathne
- Department of Veterinary Pathology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chang-Seob Seo
- K-herb Research Center, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Ji-Won Song
- Department of Veterinary Pathology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hee-Seon Park
- Department of Veterinary Pathology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Og-Sung Moon
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Republic of Korea
| | - Young-Suk Won
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Republic of Korea
| | - Hyo-Jung Kwon
- Department of Veterinary Pathology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hwa-Young Son
- Department of Veterinary Pathology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
39
|
Barnes PJ. Kinases as Novel Therapeutic Targets in Asthma and Chronic Obstructive Pulmonary Disease. Pharmacol Rev 2017; 68:788-815. [PMID: 27363440 DOI: 10.1124/pr.116.012518] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Multiple kinases play a critical role in orchestrating the chronic inflammation and structural changes in the respiratory tract of patients with asthma and chronic obstructive pulmonary disease (COPD). Kinases activate signaling pathways that lead to contraction of airway smooth muscle and release of inflammatory mediators (such as cytokines, chemokines, growth factors) as well as cell migration, activation, and proliferation. For this reason there has been great interest in the development of kinase inhibitors as anti-inflammatory therapies, particular where corticosteroids are less effective, as in severe asthma and COPD. However, it has proven difficult to develop selective kinase inhibitors that are both effective and safe after oral administration and this has led to a search for inhaled kinase inhibitors, which would reduce systemic exposure. Although many kinases have been implicated in inflammation and remodeling of airway disease, very few classes of drug have reached the stage of clinical studies in these diseases. The most promising drugs are p38 MAP kinases, isoenzyme-selective PI3-kinases, Janus-activated kinases, and Syk-kinases, and inhaled formulations of these drugs are now in development. There has also been interest in developing inhibitors that block more than one kinase, because these drugs may be more effective and with less risk of losing efficacy with time. No kinase inhibitors are yet on the market for the treatment of airway diseases, but as kinase inhibitors are improved from other therapeutic areas there is hope that these drugs may eventually prove useful in treating refractory asthma and COPD.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
40
|
Wang R, Xiao X, Shen Z, Cao L, Cao Y. Airborne fine particulate matter causes murine bronchial hyperreactivity via MAPK pathway-mediated M 3 muscarinic receptor upregulation. ENVIRONMENTAL TOXICOLOGY 2017; 32:371-381. [PMID: 26916448 DOI: 10.1002/tox.22241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 01/24/2016] [Indexed: 06/05/2023]
Abstract
Regarding the human health effects, airborne fine particulate matter 2.5 (PM2.5 ) is an important environmental risk factor. However, the underlying molecular mechanisms are largely unknown. The present study examined the hypothesis that PM2.5 causes bronchial hyperreactivity by upregulated muscarinic receptors via the mitogen-activated protein kinase (MAPK) pathway. The isolated rat bronchi segments were cultured with different concentration of PM2.5 for different time. The contractile response of the bronchi segments were recorded by a sensitive myograph. The mRNA and protein expression levels of M3 muscarinic receptors were studied by quantitative real-time PCR and immunohistochemistry, respectively. The muscarinic receptors agonist, carbachol induced a remarkable contractile response on fresh and DMSO cultured bronchial segments. Compared with the fresh or DMSO culture groups, 1.0 µg/mL of PM2.5 cultured for 24 h significantly enhanced muscarinic receptor-mediated contractile responses in bronchi with a markedly increased maximal contraction. In addition, the expression levels of mRNA and protein for M3 muscarinic receptors in bronchi of PM2.5 group were higher than that of fresh or DMSO culture groups. SB203580 (p38 inhibitor) and U0126 (MEK1/2 inhibitor) significantly inhibited the PM2.5 -induced enhanced contraction and increased mRNA and protein expression of muscarinic receptors. However, JNK inhibitor SP600125 had no effect on PM2.5 -induced muscarinic receptor upregulation and bronchial hyperreactivity. In conclusion, airborne PM2.5 upregulates muscarinic receptors, which causes subsequently bronchial hyperreactivity shown as enhanced contractility in bronchi. This process may be mediated by p38 and MEK1/2 MAPK pathways. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 371-381, 2017.
Collapse
Affiliation(s)
- Rong Wang
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Department of Pharmacy, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xue Xiao
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Zhenxing Shen
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lei Cao
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Yongxiao Cao
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| |
Collapse
|
41
|
Fei X, Zhang X, Zhang GQ, Bao WP, Zhang YY, Zhang M, Zhou X. Cordycepin inhibits airway remodeling in a rat model of chronic asthma. Biomed Pharmacother 2017; 88:335-341. [PMID: 28119235 DOI: 10.1016/j.biopha.2017.01.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/03/2017] [Accepted: 01/03/2017] [Indexed: 12/14/2022] Open
Abstract
The potential suppression role of cordycepin (Cor) on airway remodeling in a rat model of chronic asthma was investigated in this paper. We evaluated the anti-remodeling of Cor (50mg/kg) combined with or without budesonide (BUD) and investigated the possible underlying molecular mechanisms. We found that Cor attenuated immunoglobulin (Ig) E, alleviated the airway wall thickness, and decreased eosinophils and neutrophils in the bronchoalveolar lavage fluid (BALF). Notably, Cor reduced the up-regulation of IL-5, IL-13 and TNF-α in the BALF. Cor also regulated the increase of A2AARmRNA and the decrease of TGF-β1 expression. Furthermore, Cor markedly blocked p38MAPK signaling pathway activation in the OVA-driven asthmatic mice. The combination treatment of Cor and BUD showed profound efficacy in regulating the levels of inflammatory cells and the expression of IL-13, TGF-β1 and A2AARmRNA. Collectively, this study demonstrated that Cor combined with glucocorticoids treatment shows synergistically profound efficacy in inhibiting airway remodeling, and some benefits of Cor may result from the increased A2AARmRNA expression, the reduced TGF-β1 levels and the inhibition of Th2-cytokines through the suppression of the p38MAPK signaling pathways.
Collapse
Affiliation(s)
- Xia Fei
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Xue Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Guo-Qing Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Wu-Ping Bao
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Ying-Ying Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China
| | - Min Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China.
| | - Xin Zhou
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, No. 100, Haining Road, Shanghai, 200080, China.
| |
Collapse
|
42
|
Oligonucleotide Therapy for Obstructive and Restrictive Respiratory Diseases. Molecules 2017; 22:molecules22010139. [PMID: 28106744 PMCID: PMC6155767 DOI: 10.3390/molecules22010139] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/05/2017] [Accepted: 01/08/2017] [Indexed: 12/21/2022] Open
Abstract
Inhaled oligonucleotide is an emerging therapeutic modality for various common respiratory diseases, including obstructive airway diseases like asthma and chronic obstructive pulmonary disease (COPD) and restrictive airway diseases like idiopathic pulmonary fibrosis (IPF). The advantage of direct accessibility for oligonucleotide molecules to the lung target sites, bypassing systemic administration, makes this therapeutic approach promising with minimized potential systemic side effects. Asthma, COPD, and IPF are common chronic respiratory diseases, characterized by persistent airway inflammation and dysregulated tissue repair and remodeling, although each individual disease has its unique etiology. Corticosteroids have been widely prescribed for the treatment of asthma, COPD, and IPF. However, the effectiveness of corticosteroids as an anti-inflammatory drug is limited by steroid resistance in severe asthma, the majority of COPD cases, and pulmonary fibrosis. There is an urgent medical need to develop target-specific drugs for the treatment of these respiratory conditions. Oligonucleotide therapies, including antisense oligonucleotide (ASO), small interfering RNA (siRNA), and microRNA (miRNA) are now being evaluated both pre-clinically and clinically as potential therapeutics. The mechanisms of action of ASO and siRNA are highly target mRNA specific, ultimately leading to target protein knockdown. miRNA has both biomarker and therapeutic values, and its knockdown by a miRNA antagonist (antagomir) has a broader but potentially more non-specific biological outcome. This review will compile the current findings of oligonucleotide therapeutic targets, verified in various respiratory disease models and in clinical trials, and evaluate different chemical modification approaches to improve the stability and potency of oligonucleotides for the treatment of respiratory diseases.
Collapse
|
43
|
New Anti-inflammatory Drugs for COPD: Is There a Possibility of Developing Drugs That Can Fundamentally Suppress Inflammation? RESPIRATORY DISEASE SERIES: DIAGNOSTIC TOOLS AND DISEASE MANAGEMENTS 2017. [DOI: 10.1007/978-981-10-0839-9_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
44
|
Li L, Piao H, Zheng M, Jin Z, Zhao L, Yan G. Sesamin attenuates allergic airway inflammation through the suppression of nuclear factor-kappa B activation. Exp Ther Med 2016; 12:4175-4181. [PMID: 28105144 DOI: 10.3892/etm.2016.3903] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/05/2016] [Indexed: 12/30/2022] Open
Abstract
The aim of the present study is to determine the role of sesamin, the most abundant lignan in sesame seed oil, on the regulation of allergic airway inflammation in a murine asthma model. A BALB/c mouse model with allergic asthma was used to evaluate the effects of sesamin on nuclear factor-kappa B (NF-κB) activation. An enzyme-linked immunosorbent assay was used to determine protein expression in bronchoalveolar lavage (BAL) fluids. Hematoxylin and eosin staining was performed to examine histological changes. Moreover, western blot analysis was used to detect the expression of proteins in tissues. Prior to administering sesamin, the mice developed the following pathophysiological features of asthma: An increase in the number of inflammatory cells, increased levels of interleukin (IL)-4, IL-5 and IL-13, decreased levels of interferon-γ in BAL fluids and lung tissues, increased immunoglobulin E (IgE) levels in the serum and an increased activation of NF-κB in lung tissues. Following treatment with sesamin, the mice had evidently reduced peribronchiolar inflammation and airway inflammatory cell recruitment, inhibited production of several cytokines in BAL fluids and lung tissues, and decreased IgE levels. Following inhalation of ovalbumin, the administration of sesamin also inhibited the activation of NF-κB. In addition, sesamin administration reduced the phosphorylation of p38 mitogen-activated protein kinases (MAPKs). The present study demonstrates that sesamin decreases the activation of NF-κB in order to attenuate allergic airway inflammation in a murine model of asthma, possibly via the regulation of phosphorylation of p38 MAPK. These observations provide an important molecular mechanism for the potential use of sesamin in preventing and/or treating asthma, as well as other airway inflammatory disorders.
Collapse
Affiliation(s)
- Liangchang Li
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Hongmei Piao
- Department of Respiratory Medicine, Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Mingyu Zheng
- Department of Respiratory Medicine, College of Pharmacy, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Zhewu Jin
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Liguang Zhao
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Guanghai Yan
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| |
Collapse
|
45
|
Abstract
The discovery of an ever-expanding plethora of coding and non-coding RNAs with nodal and causal roles in the regulation of lung physiology and disease is reinvigorating interest in the clinical utility of the oligonucleotide therapeutic class. This is strongly supported through recent advances in nucleic acids chemistry, synthetic oligonucleotide delivery and viral gene therapy that have succeeded in bringing to market at least three nucleic acid-based drugs. As a consequence, multiple new candidates such as RNA interference modulators, antisense, and splice switching compounds are now progressing through clinical evaluation. Here, manipulation of RNA for the treatment of lung disease is explored, with emphasis on robust pharmacological evidence aligned to the five pillars of drug development: exposure to the appropriate tissue, binding to the desired molecular target, evidence of the expected mode of action, activity in the relevant patient population and commercially viable value proposition.
Collapse
|
46
|
Li LC, Piao HM, Zheng MY, Lin ZH, Choi YH, Yan GH. Ginsenoside Rh2 attenuates allergic airway inflammation by modulating nuclear factor-κB activation in a murine model of asthma. Mol Med Rep 2016; 12:6946-54. [PMID: 26502836 DOI: 10.3892/mmr.2015.4272] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 07/21/2015] [Indexed: 11/05/2022] Open
Abstract
Allergic asthma is a chronic inflammatory disease that is regulated by coordination of T-helper type 2 cell cytokines and inflammatory signaling molecules. Ginsenoside Rh2 (G-Rh2) is an active component of ginseng with anti-inflammatory and anti-tumor effects. The aim of the present study was to determine the inhibitory effects of G-Rh2 on allergic airway inflammation in a murine model of asthma, in which mice develop the following pathophysiological features of asthma: Increased abundance of inflammatory cells; increased levels of interleukin-4 (IL-4), IL-5 and IL-13; decreased abundance of interferon gamma in the bronchoalveolar lavage fluid and lung tissue; increased total and ovalbumin (OVA)-specific immunoglobulin E (IgE) levels in the serum; increased airway hyperresponsiveness (AHR); and activation of nuclear factor kappa B (NF-κB) in lung tissue. In the asthmatic mice, administration of G-Rh2 markedly reduced peribronchiolar inflammation, recruitment of airway inflammatory cells, cytokine production, total and OVA-specific IgE levels and AHR. G-Rh2 administration inhibited NF-κB activation and p38 mitogen-activated protein kinase (MAPK) phosphorylation induced by OVA inhalation. These results suggested that G-Rh2 attenuates allergic airway inflammation by regulating NF-κB activation and p38 MAPK phosphorylation. The present study identified the molecular mechanisms of action of G-Rh2, which supported the potential use of G-Rh2 to prevent and/or treat asthma and other airway inflammatory disorders.
Collapse
|
47
|
Wang R, Xiao X, Cao L, Shen ZX, Lei Y, Cao YX. Airborne fine particulate matter induces an upregulation of endothelin receptors on rat bronchi. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2016; 209:11-20. [PMID: 26618262 DOI: 10.1016/j.envpol.2015.10.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/23/2015] [Indexed: 06/05/2023]
Abstract
Airborne fine particulate matter (PM2.5) is a risk factor for respiratory diseases. However, little is known about the effects of PM2.5 on bronchi. The present study investigated the effect of airborne PM2.5 on rat bronchi and the underlying mechanisms. Isolated rat bronchial segments were cultured for 24 h. Endothelin (ET) receptor-mediated contractile responses were recorded using a wire myograph. The mRNA and protein expression levels of ET receptors were studied using quantitative real-time PCR, Western blotting, and immunohistochemistry. The results demonstrated that ETA and ETB receptor agonists induced remarkable contractile responses on fresh and cultured bronchial segments. PM2.5 (1.0 or 3.0 μg/ml) significantly enhanced ETA and ETB receptor-mediated contractile responses in bronchi with a markedly increased maximal contraction compared to the DMSO or fresh groups. PM2.5 increased the mRNA and protein expression levels of ETA and ETB receptors. U0126 (a MEK1/2 inhibitor) and SB203580 (a p38 inhibitor) significantly suppressed PM2.5-induced increases in ETB receptor-mediated contractile responses, mRNA and protein levels. SP600125 (a JNK inhibitor) and SB203580 significantly abrogated the PM2.5-induced enhancement of ETA receptor-mediated contraction and receptor expression. In conclusion, PM2.5 upregulates ET receptors in bronchi. ETB receptor upregulation is associated with MEK1/2 and p38 pathways, and the upregulation of ETA receptor is involved in JNK and p38 pathways.
Collapse
Affiliation(s)
- Rong Wang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xue Xiao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Lei Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
| | - Zhen-xing Shen
- Department of Environmental Science and Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ying Lei
- Department of Pharmacy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yong-xiao Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
| |
Collapse
|
48
|
meso-Dihydroguaiaretic acid attenuates airway inflammation and mucus hypersecretion in an ovalbumin-induced murine model of asthma. Int Immunopharmacol 2016; 31:239-47. [PMID: 26773771 DOI: 10.1016/j.intimp.2015.12.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 12/09/2015] [Accepted: 12/22/2015] [Indexed: 01/01/2023]
Abstract
meso-Dihydroguaiaretic acid (MDGA), which is a dibenzylbutane lignin isolated from the ethyl acetate fraction of Saururus chinensis, has various biological activities, including anti-oxidative, anti-inflammatory, anti-bacterial, and neuroprotective effects. However, no report has examined the potential anti-asthmatic activity of MDGA. In this study, we evaluated the protective effects of MDGA on asthmatic responses, particularly airway inflammation and mucus hypersecretion in an ovalbumin (OVA)-induced murine model of asthma. Intragastric administration of MDGA significantly lowered the productions of interleukin (IL)-4, IL-5, IL-13, tumor necrosis-α (TNF-α), eotaxin, monocyte chemoattractant protein-1 (MCP-1), vascular cell adhesion molecule-1 (VCAM-1), and immunoglobulin (Ig)E in bronchoalveolar lavage fluid (BALF), plasma, or lung tissues. Histological studies showed that MDGA inhibited OVA-induced inflammatory cell infiltration and mucus production in the respiratory tract. Moreover, MDGA markedly attenuated the OVA-induced activations of nuclear factor kappa B (NF-κB), extracellular-signal-regulated kinases 1/2 (ERK1/2), and p38 mitogen-activated protein kinase (p38 MAPK). Together, these results suggest that MDGA effectively inhibits airway inflammation and mucus hypersecretion by downregulating the levels of T helper 2 (Th2) cytokines, chemokines, and adhesion molecules, and inhibiting the activations of NF-κB and MAPKs.
Collapse
|
49
|
Barnes PJ. Therapeutic approaches to asthma-chronic obstructive pulmonary disease overlap syndromes. J Allergy Clin Immunol 2015; 136:531-45. [PMID: 26343937 DOI: 10.1016/j.jaci.2015.05.052] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 12/14/2022]
Abstract
The recognition that there are some patients with features of asthma and chronic obstructive pulmonary disease (COPD) has highlighted the need to develop more specific treatments for these clinical phenotypes. Some patients with COPD have predominantly eosinophilic inflammation and might respond to high doses of inhaled corticosteroids and newly developed specific antieosinophil therapies, including blocking antibodies against IL-5, IL-13, IL-33, and thymic stromal lymphopoietin, as well as oral chemoattractant receptor-homologous molecule expressed on TH2 cells antagonists. Other patients have severe asthma or are asthmatic patients who smoke with features of COPD-induced inflammation and might benefit from treatments targeting neutrophils, including macrolides, CXCR2 antagonists, phosphodiesterase 4 inhibitors, p38 mitogen-activating protein kinase inhibitors, and antibodies against IL-1 and IL-17. Other patients appear to have largely fixed obstruction with little inflammation and might respond to long-acting bronchodilators, including long-acting muscarinic antagonists, to reduce hyperinflation. Highly selected patients with severe asthma might benefit from bronchial thermoplasty. Some patients with overlap syndromes can be conveniently treated with triple fixed-dose combination inhaler therapy with an inhaled corticosteroid, long-acting β2-agonist, and long-acting muscarinic antagonist, several of which are now in development. Corticosteroid resistance is a feature of asthma-COPD overlap syndrome, and understanding the various molecular mechanisms of this resistance has identified novel therapeutic targets and presented the prospect of therapies that can restore corticosteroid responsiveness.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom.
| |
Collapse
|
50
|
Li F, Chen Y, Zhang Z, Ouyang J, Wang Y, Yan R, Huang S, Gao GF, Guo G, Chen JL. Robust expression of vault RNAs induced by influenza A virus plays a critical role in suppression of PKR-mediated innate immunity. Nucleic Acids Res 2015; 43:10321-37. [PMID: 26490959 PMCID: PMC4666359 DOI: 10.1093/nar/gkv1078] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/07/2015] [Indexed: 12/29/2022] Open
Abstract
Protein kinase R (PKR) is a vital component of host innate immunity against viral infection. However, the mechanism underlying inactivation of PKR by influenza A virus (IAV) remains elusive. Here, we found that vault RNAs (vtRNAs) were greatly induced in A549 cells and mouse lungs after infection with IAV. The viral NS1 protein was shown to be the inducer triggering the upregulation of vtRNAs. Importantly, silencing vtRNA in A549 cells significantly inhibited IAV replication, whereas overexpression of vtRNAs markedly promoted the viral replication. Furthermore, in vivo studies showed that disrupting vtRNA expression in mice significantly decreased IAV replication in infected lungs. The vtRNA knockdown animals exhibited significantly enhanced resistance to IAV infection, as evidenced by attenuated acute lung injury and spleen atrophy and consequently increased survival rates. Interestingly, vtRNAs promoted viral replication through repressing the activation of PKR and the subsequent antiviral interferon response. In addition, increased expression of vtRNAs was required for efficient suppression of PKR by NS1 during IAV infection. Moreover, vtRNAs were also significantly upregulated by infections of several other viruses and involved in the inactivation of PKR signaling by these viruses. These results reveal a novel mechanism by which some viruses circumvent PKR-mediated innate immunity.
Collapse
Affiliation(s)
- Fang Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China
| | - Yuhai Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China
| | - Zhaoyuan Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China School of Life Sciences, Anhui University, Hefei 230601, China
| | - Jing Ouyang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China
| | - Yi Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China
| | - Ruoxiang Yan
- College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - George Fu Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China
| | - Guijie Guo
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China
| | - Ji-Long Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|