1
|
Rao R, Mei J, Chen H, Yang C. Association of metformin use with asthma development and adverse outcomes: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e39785. [PMID: 39465742 PMCID: PMC11460891 DOI: 10.1097/md.0000000000039785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Asthma and diabetes are prevalent chronic diseases affecting a significant population globally. Research has suggested that metformin, a commonly used medication for diabetes management, may also have beneficial effects in enhancing asthma outcomes. Considering the comorbidity of asthma and diabetes, a comprehensive analysis was performed to investigate the efficacy of metformin in reducing adverse outcomes of asthma patients with diabetes. METHODS To gather relevant data, we conducted a systematic search of the PubMed, Embase, and CENTRAL databases for observational studies published prior to September 2023. We specifically looked for studies involving individuals diagnosed with both asthma and diabetes, comparing the incidence and severity of asthma exacerbations in metformin users versus nonusers. The inclusion criteria encompassed studies that recruited participants aged 18 years and older. The primary outcome of interest was the risk of newly developing asthma, while secondary outcomes included the adjusted risk of asthma-induced exacerbations, emergency room visits, and hospitalizations. All data analyses and visualizations were performed using the R programming language. RESULTS We identified and included 7 studies involving a total of 1,176,398 patients in our analysis. The pooled effect size indicated a potential reduction in the incidence of newly developed asthma among patients with type 2 diabetes who used metformin, although this finding did not reach statistical significance. Similar conclusions have also been observed in other outcomes, such as exacerbation, asthma-related emergency department visits, risk of systemic corticosteroid prescription. The only positive outcome is that the use of metformin can reduce the chance of patients being hospitalized due to asthma. CONCLUSION In most outcome indicators, it cannot be assumed that the use of metformin can reduce asthma-related adverse events. However, the conclusion is not so certain, and longer observation and more evidence are still required. Metformin still shows some potential in the intervention of respiratory diseases.
Collapse
Affiliation(s)
- Rui Rao
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Juan Mei
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Hudie Chen
- Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Chuanjing Yang
- Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
2
|
Zhang J, Shimozaki K, Hattori S, Pastukh V, Maloney D, Hogan MV, Wang JHC. Metformin lotion promotes scarless skin tissue formation through AMPK activation, TGF-β1 inhibition, and reduced myofibroblast numbers. PLoS One 2024; 19:e0311147. [PMID: 39331598 PMCID: PMC11433050 DOI: 10.1371/journal.pone.0311147] [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: 07/22/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024] Open
Abstract
Scar tissue formation following skin wound healing is a challenging public health problem. Skin regeneration and preventing the formation of scar tissue by currently available commercial products are largely ineffective. This study aimed to test the efficacy of a novel topical metformin lotion (ML) in inhibiting scar tissue formation during skin wound healing in rats and to determine the mechanisms of action involved. A 6% ML was prepared in our laboratory. A skin wound healing model in rats was used. The wounded rats were divided into two groups and treated daily for 10 days as follows: Group 1 received a daily application of 50 mg of control lotion, or 0% ML (totaling 100 mg of lotion per rat), and Group 2 received a daily application of 50 mg of 6% ML (totaling 100 mg of 6% ML per rat). Blood samples from the heart of each rat were analyzed for inflammatory markers, HMGB1 and IL-1β, using ELISA, and immunological and histological analyses were performed on skin tissue sections. ML decreased levels of inflammatory markers HMGB1 and IL-1β in the serum of rats and inhibited the release of HMGB1 from cell nuclei into the skin tissue matrix. Additionally, ML demonstrated anti-fibrotic properties by enhancing AMPK activity, decreasing the expression of TGF-β1, reducing the number of myofibroblasts, decreasing the production of collagen III, and increasing the expression of collagen I. ML promotes the regeneration of high-quality skin during wound healing by reducing scar tissue formation. This effect is mediated through the activation of AMPK, inhibition of TGF-β1, and a decrease in the number of myofibroblasts.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kengo Shimozaki
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Soichi Hattori
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Vasyl Pastukh
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Derek Maloney
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - MaCalus V. Hogan
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James H-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
3
|
Narendra DK, Khurana S. Asthma and Hyperglycemia: Exploring the Interconnected Pathways. Diagnostics (Basel) 2024; 14:1869. [PMID: 39272654 PMCID: PMC11393853 DOI: 10.3390/diagnostics14171869] [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: 06/18/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
The interplay between asthma and glucose metabolism disorders, such as hyperglycemia, has gained increasing attention due to the potential exacerbation of asthma symptoms and severity. This review explores the complex relationship between hyperglycemia and asthma, emphasizing the pathophysiological links, the impact of glucose metabolism disorders on asthma, and the effects of asthma medications on glucose levels. Hyperglycemia, often induced by asthma treatments like corticosteroids, has been associated with an increased risk of asthma exacerbations. This review delves into the pathophysiology underlying this association, highlighting the role of insulin resistance, metabolic syndrome, and obesity in both the development and management of asthma. Metabolic syndrome, characterized by abdominal obesity and hyperglycemia, independently increases the risk of worsening respiratory symptoms and asthma. Furthermore, this review examines the influence of various antidiabetic medications on asthma outcomes. Biguanides, like metformin, have shown promise in improving asthma outcomes in patients with type 2 diabetes mellitus and asthma. However, other medications have mixed results regarding their impact on asthma control and lung function. Considering these findings, this review advocates for further research into the role of metabolic pathways in asthma management. It calls for comparative studies and the inclusion of asthma-related outcomes in clinical trials of antidiabetic drugs to better understand their potential benefits for individuals with obesity and concurrent asthma.
Collapse
Affiliation(s)
| | - Sandhya Khurana
- University of Rochester Medical Center, Rochester, NY 14620, USA
| |
Collapse
|
4
|
Kimura Y, Jo T, Inoue N, Suzukawa M, Hashimoto Y, Kumazawa R, Ishimaru M, Matsui H, Yokoyama A, Tanaka G, Sasabuchi Y, Yasunaga H. Association of Novel Antihyperglycemic Drugs Versus Metformin With a Decrease in Asthma Exacerbations. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:2035-2044. [PMID: 38734374 DOI: 10.1016/j.jaip.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/10/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Similar to metformin, dipeptidyl peptidase-4 inhibitors (DPP-4 Is), glucagon-like peptidase 1 receptor agonists (GLP-1 RAs), and sodium glucose co-transporter-2 inhibitors (SGLT-2 Is) may improve control of asthma owing to their multiple potential mechanisms, including differential improvements in glycemic control, direct anti-inflammatory effects, and systemic changes in metabolism. OBJECTIVE To investigate whether these novel antihyperglycemic drugs were associated with fewer asthma exacerbations compared with metformin in patients with asthma comorbid with type 2 diabetes. METHODS Using a Japanese national administrative database, we constructed 3 active comparators-new user cohorts of 137,173 patients with a history of asthma starting the novel antihyperglycemic drugs and metformin between 2014 and 2022. Patient characteristics were balanced using overlap propensity score weighting. The primary outcome was the first exacerbation requiring systemic corticosteroids, and the secondary outcomes included the number of exacerbations requiring systemic corticosteroids. RESULTS DPP-4 Is and GLP-1 RAs were associated with a higher incidence of exacerbations requiring systemic corticosteroids compared with metformin (DPP-4 Is: 18.2 vs 17.4 per 100 person-years, hazard ratio: 1.09, 95% confidence interval [CI]: 1.05-1.14; GLP-1 RAs: 24.9 vs 19.0 per 100 person-years, hazard ratio: 1.14, 95% CI: 1.01-1.28). In contrast, the incidence of exacerbations requiring systemic corticosteroids was similar between the SGLT-2 Is and metformin groups (17.3 vs 18.1 per 100 person-years, hazard ratio: 1.00, 95% CI: 0.97-1.03). While DPP-4 Is and GLP-1 RAs were associated with more exacerbations requiring systemic corticosteroids, SGLT-2 Is were associated with slightly fewer exacerbations requiring systemic corticosteroids (53.7 vs 56.6 per 100 person-years, rate ratio: 0.95, 95% CI: 0.91-0.99). CONCLUSIONS While DPP-4 Is and GLP-1 RAs were associated with poorer control of asthma compared with metformin, SGLT-2 Is offered asthma control comparable to that of metformin.
Collapse
Affiliation(s)
- Yuya Kimura
- Department of Clinical Epidemiology and Health Economics, School of Public Health, the University of Tokyo, Tokyo, Japan; Clinical Research Center, National Hospital Organization Tokyo Hospital, Tokyo, Japan.
| | - Taisuke Jo
- Department of Health Services Research, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan; Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Norihiko Inoue
- Department of Health Policy and Informatics, Tokyo Medical and Dental University Graduate School, Tokyo, Japan; Department of Clinical Data Management and Research, Clinical Research Center, National Hospital Organization Headquarters, Tokyo, Japan
| | - Maho Suzukawa
- Clinical Research Center, National Hospital Organization Tokyo Hospital, Tokyo, Japan
| | - Yohei Hashimoto
- Save Sight Institute, the University of Sydney, Sydney, NSW, Australia
| | - Ryosuke Kumazawa
- Department of Clinical Epidemiology and Health Economics, School of Public Health, the University of Tokyo, Tokyo, Japan
| | - Miho Ishimaru
- Department of Clinical Epidemiology and Health Economics, School of Public Health, the University of Tokyo, Tokyo, Japan; Department of Health Services Research, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroki Matsui
- Department of Clinical Epidemiology and Health Economics, School of Public Health, the University of Tokyo, Tokyo, Japan
| | - Akira Yokoyama
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Goh Tanaka
- Department of Respiratory Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Yusuke Sasabuchi
- Department of Real-world Evidence, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public Health, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Lei J, Shu Z, Zhu H, Zhao L. AMPK Regulates M1 Macrophage Polarization through the JAK2/STAT3 Signaling Pathway to Attenuate Airway Inflammation in Obesity-Related Asthma. Inflammation 2024:10.1007/s10753-024-02070-x. [PMID: 38886294 DOI: 10.1007/s10753-024-02070-x] [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: 04/07/2024] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
Abstract-Obesity-related asthma is primarily characterized by nonallergic inflammation, with pathogenesis involving oxidative stress, metabolic imbalance, and immunoinflammatory mechanisms. M1 macrophages, which predominantly secrete pro-inflammatory factors, mediate insulin resistance and systemic metabolic inflammation in obese individuals. Concurrently, adenosine monophosphate-activated protein kinase (AMPK) serves as a critical regulator of intracellular energy metabolism and is closely associated with macrophage activation. However, their specific roles and associated mechanisms in obesity-related asthma remain to be explored. In this study, we investigated the macrophage polarization status and potential interventional mechanisms through obesity-related asthmatic models and lipopolysaccharide (LPS) -treated RAW264.7 cell with a comprehensive series of evaluations, including HE, PAS and Masson staining of lung histopathology, immunohistochemical staining, immunofluorescence technology, qRT-PCR, Western Blot, and ELISA inflammatory factor analysis. The results revealed M1 macrophage polarization in obesity-related asthmatic lung tissue alongside downregulation of AMPK expression. Under LPS stimulation, exogenous AMPK activation attenuated M1 macrophage polarization via the Janus kinase 2/ signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway. Additionally, in obesity-related asthmatic mice, AMPK activation was found to alleviate airway inflammation by regulating M1 macrophage polarization, the mechanism closely associated with the JAK2/STAT3 pathway. These findings not only advance our understanding of macrophage polarization in obesity-related asthma, but also provide new therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jiahui Lei
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| | - Zhenhui Shu
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, 450003, China
| | - He Zhu
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, 450003, China
| | - Limin Zhao
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450003, China.
- To whom correspondence should be addressed at Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan Province, China.
| |
Collapse
|
6
|
Liu T, Wei H, Zhang L, Ma C, Wei Y, Jiang T, Li W. Metformin attenuates lung ischemia-reperfusion injury and necroptosis through AMPK pathway in type 2 diabetic recipient rats. BMC Pulm Med 2024; 24:237. [PMID: 38745191 PMCID: PMC11094932 DOI: 10.1186/s12890-024-03056-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Diabetes mellitus (DM) can aggravate lung ischemia-reperfusion (I/R) injury and is a significant risk factor for recipient mortality after lung transplantation. Metformin protects against I/R injury in a variety of organs. However, the effect of metformin on diabetic lung I/R injury remains unclear. Therefore, this study aimed to observe the effect and mechanism of metformin on lung I/R injury following lung transplantation in type 2 diabetic rats. METHODS Sprague-Dawley rats were randomly divided into the following six groups: the control + sham group (CS group), the control + I/R group (CIR group), the DM + sham group (DS group), the DM + I/R group (DIR group), the DM + I/R + metformin group (DIRM group) and the DM + I/R + metformin + Compound C group (DIRMC group). Control and diabetic rats underwent the sham operation or left lung transplantation operation. Lung function, alveolar capillary permeability, inflammatory response, oxidative stress, necroptosis and the p-AMPK/AMPK ratio were determined after 24 h of reperfusion. RESULTS Compared with the CIR group, the DIR group exhibited decreased lung function, increased alveolar capillary permeability, inflammatory responses, oxidative stress and necroptosis, but decreased the p-AMPK/AMPK ratio. Metformin improved the function of lung grafts, decreased alveolar capillary permeability, inflammatory responses, oxidative stress and necroptosis, and increased the p-AMPK/AMPK ratio. In contrast, the protective effects of metformin were abrogated by Compound C. CONCLUSIONS Metformin attenuates lung I/R injury and necroptosis through AMPK pathway in type 2 diabetic lung transplant recipient rats.
Collapse
Affiliation(s)
- Tianhua Liu
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Hong Wei
- Department of Anesthesiology, Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lijuan Zhang
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Can Ma
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Yuting Wei
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Tao Jiang
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Wenzhi Li
- Department of Anesthesiology, Hei Long Jiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, NO.246, Xuefu Road, Nangang District, Harbin, Heilongjiang Province, 150081, China.
| |
Collapse
|
7
|
Xuan L, Yang S, Ren L, Liu H, Zhang W, Sun Y, Xu B, Gong L, Liu L. Akebia saponin D attenuates allergic airway inflammation through AMPK activation. J Nat Med 2024; 78:393-402. [PMID: 38175326 DOI: 10.1007/s11418-023-01762-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/10/2023] [Indexed: 01/05/2024]
Abstract
Akebia saponin D (ASD) is a bioactive triterpenoid saponin extracted from Dipsacus asper Wall. ex DC.. This study aimed to investigate the effects of ASD on allergic airway inflammation. Human lung epithelial BEAS-2B cells and bone marrow-derived mast cells (BMMCs) were pretreated with ASD (50, 100 and 200 μΜ) and AMPK activator 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) (1 mM), and then stimulated with lipopolysaccharide (LPS) or IL-33. Pretreatment with ASD and AICAR significantly inhibited TNF-α and IL-6 production from BEAS-2B cells, and IL-13 production from BMMCs. Moreover, pretreatment with ASD and AICAR significantly increased p-AMPK expression in BEAS-2B cells. Inhibition of AMPK by siRNA and compound C partly abrogated the suppression effect of ASD on TNF-α, IL-6, and IL-13 production. Asthma murine model was induced by ovalbumin (OVA) challenge and treated with ASD (150 and 300 mg/kg) or AICAR (100 mg/kg). Infiltration of eosinophils, neutrophils, monocytes, and lymphocytes, and production of TNF-α, IL-6, IL-4, and IL-13 were attenuated in ASD and AICAR treated mice. Lung histopathological changes were also ameliorated after ASD and AICAR treatment. Additionally, it showed that treatment with ASD and AICAR increased p-AMPK expression in the lung tissues. In conclusion, ASD exhibited protective effects on allergic airway inflammation through the induction of AMPK activation.
Collapse
Affiliation(s)
- Lingling Xuan
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China.
| | - Song Yang
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China
| | - Lulu Ren
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China
| | - He Liu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China
| | - Wen Zhang
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China
| | - Yuan Sun
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China
| | - Benshan Xu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China
| | - Lili Gong
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China.
| | - Lihong Liu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Beijing, 100020, China.
| |
Collapse
|
8
|
Bartziokas K, Papaioannou AI, Drakopanagiotakis F, Gouveri E, Papanas N, Steiropoulos P. Unraveling the Link between Ιnsulin Resistance and Bronchial Asthma. Biomedicines 2024; 12:437. [PMID: 38398039 PMCID: PMC10887139 DOI: 10.3390/biomedicines12020437] [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: 01/13/2024] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Evidence from large epidemiological studies has shown that obesity may predispose to increased Th2 inflammation and increase the odds of developing asthma. On the other hand, there is growing evidence suggesting that metabolic dysregulation that occurs with obesity, and more specifically hyperglycemia and insulin resistance, may modify immune cell function and in some degree systemic inflammation. Insulin resistance seldom occurs on its own, and in most cases constitutes a clinical component of metabolic syndrome, along with central obesity and dyslipidemia. Despite that, in some cases, hyperinsulinemia associated with insulin resistance has proven to be a stronger risk factor than body mass in developing asthma. This finding has been supported by recent experimental studies showing that insulin resistance may contribute to airway remodeling, promotion of airway smooth muscle (ASM) contractility and proliferation, increase of airway hyper-responsiveness and release of pro-inflammatory mediators from adipose tissue. All these effects indicate the potential impact of hyperinsulinemia on airway structure and function, suggesting the presence of a specific asthma phenotype with insulin resistance. Epidemiologic studies have found that individuals with severe and uncontrolled asthma have a higher prevalence of glycemic dysfunction, whereas longitudinal studies have linked glycemic dysfunction to an increased risk of asthma exacerbations. Since the components of metabolic syndrome interact with one another so much, it is challenging to identify each one's specific role in asthma. This is why, over the last decade, additional studies have been conducted to determine whether treatment of type 2 diabetes mellitus affects comorbid asthma as shown by the incidence of asthma, asthma control and asthma-related exacerbations. The purpose of this review is to present the mechanism of action, and existing preclinical and clinical data, regarding the effect of insulin resistance in asthma.
Collapse
Affiliation(s)
| | - Andriana I. Papaioannou
- 1st University Department of Respiratory Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Fotios Drakopanagiotakis
- Department of Pneumonology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Evanthia Gouveri
- Diabetes Centre, 2nd Department of Internal Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.G.); (N.P.)
| | - Nikolaos Papanas
- Diabetes Centre, 2nd Department of Internal Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.G.); (N.P.)
| | - Paschalis Steiropoulos
- Department of Pneumonology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
9
|
Sun H, Wu Y, Xiong Z, Gu Y, Jia Q, Ru Z, Peng Y, Kang Z, Li Y, Huang Y, Yin S, Guo K, Feng C, Tang J, Gao Z, Wang Y, Yang X. Amphibian-derived peptide RL-RF10 ameliorates paraquat-induced pulmonary fibrosis injury. Biomed Pharmacother 2024; 171:116184. [PMID: 38244328 DOI: 10.1016/j.biopha.2024.116184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 01/22/2024] Open
Abstract
Pulmonary fibrosis is the result of dysfunctional repair after lung tissue injury, characterized by fibroblast proliferation and massive extracellular matrix aggregation. Once fibrotic lesions develop, effective treatment is difficult, with few drugs currently available. Here, we identified a short cyclic decapeptide RL-RF10 derived from frog skin secretions as a potential novel lead molecule for the amelioration of pulmonary fibrosis. In vivo experiments indicated that RL-RF10 treatment ameliorated lung histopathological damage and fibrogenesis after paraquat (PQ) induction in a concentration-dependent manner. On day 7, bronchoalveolar lavage fluid assays performed on mice showed that RL-RF10 exerted anti-inflammatory effects by decreasing the expression of inflammation-related factors, including transforming growth factor-β1 (TGF-β1) and tumor necrosis factor-α, in lung tissue. In addition, RL-RF10 down-regulated the levels of collagen I, collagen III, and vimentin, while increasing the expression of E-cadherin to inhibit epithelial-mesenchymal transition. Further research demonstrated that the SMAD2/3 signaling pathway, which is strongly linked to TGF-β1, played a critical function in enhancing the pulmonary fibrosis relief achieved by RL-RF10. Both in vivo and in vitro assays showed that RL-RF10 treatment led to a significant reduction in the phosphorylation levels of SMAD2 and SMAD3 following PQ induction. Overall, we investigated the protective effects and underlying mechanisms of the RL-RF10 peptide against pulmonary fibrosis and demonstrated its potential as a novel therapeutic drug candidate for the treatment of pulmonary fibrotic diseases.
Collapse
Affiliation(s)
- Huiling Sun
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yutong Wu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Ziqian Xiong
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yuanqi Gu
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Qiuye Jia
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Zeqiong Ru
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Ying Peng
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Zijian Kang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yuansheng Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yubing Huang
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Saige Yin
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Kun Guo
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Chengan Feng
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Jing Tang
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Zhenhua Gao
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China; Yunnan Province Clinical Research Center for Chronic Kidney Disease, Kunming, Yunnan, 650032, China.
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, Yunnan, 650504, China.
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, 650500, China.
| |
Collapse
|
10
|
Ararat E, Landes RD, Forno E, Tas E, Perry TT. Metformin use is associated with decreased asthma exacerbations in adolescents and young adults. Pediatr Pulmonol 2024; 59:48-54. [PMID: 37772681 PMCID: PMC10872793 DOI: 10.1002/ppul.26704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023]
Abstract
RATIONALE Metformin is a commonly used antidiabetes medication with suggested anti-inflammatory and antioxidative effects. Metformin use has been associated with lower risk of asthma exacerbations and hospitalizations in adults. Here, we aimed to evaluate how asthma exacerbation rates changed after adolescents and young adults were prescribed metformin, and to learn if those changes were related to metformin prescription adherence. METHODS Using secondary data of patients between 12 and 20 years old with asthma diagnosis and a metformin prescription from the Arkansas All Payers Claim Database and Arkansas School body mass index (BMI) database, we estimated the change in annualized asthma exacerbation rates after metformin prescription. We also evaluated the association of prescription adherence to the changes in those rates using univariate and multivariate regression models. RESULTS A total of 464 patients met inclusion criteria. Outpatient exacerbation rates decreased after metformin prescription (13.4% only before vs. 7.8% only after, p = .009), and the annualized rate decreased more after metformin prescription as adherence increased (rank r = -.165, p < .001). After adjusting for potential confounders-age, sex, BMI, and inhaled corticoid steroid use-the strength of the association was attenuated. CONCLUSIONS Asthma exacerbation rates decreased after metformin prescription, but a larger sample of patients who have experienced exacerbations and including patients with asthma who have not been prescribed metformin is needed to better know whether these decreases are driven by metformin use.
Collapse
Affiliation(s)
- Erhan Ararat
- Department of Pediatrics, Division of Pulmonology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Reid D Landes
- Department of Biostatistics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Erick Forno
- Division of Pediatric Pulmonary, Allergy, and Sleep Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Emir Tas
- Pediatric Endocrinology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Tamara T Perry
- Department of Pediatrics, Allergy and Immunology, University of Arkansas for Medical Sciences and Arkansas Children’s Research Institute, Little Rock, AR, USA
| |
Collapse
|
11
|
Zhang J, Brown R, Hogan MV, Wang JHC. Mitigating Scar Tissue Formation in Tendon Injuries: Targeting HMGB1, AMPK Activation, and Myofibroblast Migration All at Once. Pharmaceuticals (Basel) 2023; 16:1739. [PMID: 38139865 PMCID: PMC10748062 DOI: 10.3390/ph16121739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Tendon injuries, while prevalent, present significant challenges regarding their structural and functional restoration. Utilizing alpha-smooth muscle actin (α-SMA)-Ai9-scleraxis (Scx)-green fluorescent protein (GFP) transgenic mice, which exhibit both Scx (a tendon cell marker) and α-SMA (a myofibroblast marker), we explored the effects of metformin (Met) on tendon healing, repair, and its mechanisms of action. Our findings revealed that intraperitoneal (IP) injections of Met, administered before or after injury, as well as both, effectively prevented the release of HMGB1 into the tendon matrix and reduced circulating levels of HMGB1. Additionally, Met treatment increased and activated AMPK and suppressed TGF-β1 levels within the healing tendon. Tendon healing was also improved by blocking the migration of α-SMA+ myofibroblasts, reducing the prevalence of disorganized collagen fibers and collagen type III. It also enhanced the presence of collagen type I. These outcomes highlight Met's anti-fibrotic properties in acutely injured tendons and suggest its potential for repurposing as a therapeutic agent to minimize scar tissue formation in tendon injuries, which could have profound implications in clinical practice.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
| | - Roshawn Brown
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
| | - MaCalus V. Hogan
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - James H-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, E-1640 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA; (J.Z.); (R.B.); (M.V.H.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
12
|
Shang R, Miao J. Mechanisms and effects of metformin on skeletal muscle disorders. Front Neurol 2023; 14:1275266. [PMID: 37928155 PMCID: PMC10621799 DOI: 10.3389/fneur.2023.1275266] [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: 08/10/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Skeletal muscle disorders are mostly genetic and include several rare diseases. With disease progression, muscle fibrosis and adiposis occur, resulting in limited mobility. The long course of these diseases combined with limited treatment options affect patients both psychologically and economically, hence the development of novel treatments for neuromuscular diseases is crucial to obtain a better quality of life. As a widely used hypoglycemic drug in clinical practice, metformin not only has anti-inflammatory, autophagy-regulating, and mitochondrial biogenesis-regulating effects, but it has also been reported to improve the symptoms of neuromuscular diseases, delay hypokinesia, and regulate skeletal muscle mass. However, metformin's specific mechanism of action in neuromuscular diseases requires further elucidation. This review summarizes the evidence showing that metformin can regulate inflammation, autophagy, and mitochondrial biogenesis through different pathways, and further explores its mechanism of action in Duchenne muscular dystrophy, statin-associated muscle disorders, and age-related sarcopenia. This review clarifies the directions of future research on therapy for neuromuscular diseases.
Collapse
Affiliation(s)
| | - Jing Miao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Shailesh H, Bhat AA, Janahi IA. Obesity-Associated Non-T2 Mechanisms in Obese Asthmatic Individuals. Biomedicines 2023; 11:2797. [PMID: 37893170 PMCID: PMC10603840 DOI: 10.3390/biomedicines11102797] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/30/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Obesity and asthma are two common health issues that have shown increased prevalence in recent years and have become a significant socioeconomic burden worldwide. Obesity increases asthma incidence and severity. Obese asthmatic individuals often experience increased exacerbation rates, enhanced airway remodeling, and reduced response to standard corticosteroid therapy. Recent studies indicate that obesity-associated non-T2 factors such as mechanical stress, hyperinsulinemia, systemic inflammation, adipose tissue mediators, metabolic dysregulation, microbiome dysbiosis, and high-fat-diet are responsible for increased asthma symptoms and reduced therapeutic response in obese asthmatic individuals. This manuscript reviews the recent findings highlighting the role of obesity-associated factors that contribute to airway hyper-reactivity, airway inflammation and remodeling, and immune cell dysfunction, consequently contributing to worsening asthma symptoms. Furthermore, the review also discusses the possible future therapies that might play a role in reducing asthma symptoms by diminishing the impact of obesity-associated non-T2 factors.
Collapse
Affiliation(s)
| | - Ajaz A. Bhat
- Precision Medicine in Diabetes, Obesity and Cancer Research Program, Department of Human Genetics, Sidra Medicine, Doha 26999, Qatar;
| | - Ibrahim A. Janahi
- Department of Medical Education, Sidra Medicine, Doha 26999, Qatar;
- Department of Pediatric Medicine, Sidra Medicine, Doha 26999, Qatar
- Department of Pediatrics, Weill Cornell Medicine, Doha 24144, Qatar
| |
Collapse
|
14
|
Malaekeh-Nikouei A, Shokri-Naei S, Karbasforoushan S, Bahari H, Baradaran Rahimi V, Heidari R, Askari VR. Metformin beyond an anti-diabetic agent: A comprehensive and mechanistic review on its effects against natural and chemical toxins. Biomed Pharmacother 2023; 165:115263. [PMID: 37541178 DOI: 10.1016/j.biopha.2023.115263] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023] Open
Abstract
In addition to the anti-diabetic effect of metformin, a growing number of studies have shown that metformin has some exciting properties, such as anti-oxidative capabilities, anticancer, genomic stability, anti-inflammation, and anti-fibrosis, which have potent, that can treat other disorders other than diabetes mellitus. We aimed to describe and review the protective and antidotal efficacy of metformin against biologicals, chemicals, natural, medications, pesticides, and radiation-induced toxicities. A comprehensive search has been performed from Scopus, Web of Science, PubMed, and Google Scholar databases from inception to March 8, 2023. All in vitro, in vivo, and clinical studies were considered. Many studies suggest that metformin affects diseases other than diabetes. It is a radioprotective and chemoprotective drug that also affects viral and bacterial diseases. It can be used against inflammation-related and apoptosis-related abnormalities and against toxins to lower their effects. Besides lowering blood sugar, metformin can attenuate the effects of toxins on body weight, inflammation, apoptosis, necrosis, caspase-3 activation, cell viability and survival rate, reactive oxygen species (ROS), NF-κB, TNF-α, many interleukins, lipid profile, and many enzymes activity such as catalase and superoxide dismutase. It also can reduce the histopathological damages induced by many toxins on the kidneys, liver, and colon. However, clinical trials and human studies are needed before using metformin as a therapeutic agent against other diseases.
Collapse
Affiliation(s)
- Amirhossein Malaekeh-Nikouei
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Shokri-Naei
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sobhan Karbasforoushan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Bahari
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Heidari
- Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran; Research Center for Cancer Screening and Epidemiology, AJA University of Medical Sciences, Tehran, Iran
| | - Vahid Reza Askari
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Fu D, Zhao H, Huang Y, Li J, Feng H, Li A, Liu Y, He L. Metformin regulates the effects of IR and IGF-1R methylation on mast cell activation and airway reactivity in diabetic rats with asthma through miR-152-3p/DNMT1 axis. Cell Biol Toxicol 2023; 39:1851-1872. [PMID: 36547818 DOI: 10.1007/s10565-022-09787-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND/AIM Metformin is a drug for treating type 2 diabetes mellitus (T2DM). Recently, metformin has been shown to reduce the risks of asthma-associated outcomes and asthma deterioration, thereby holding promise as a superior medicine for diabetic patients with asthma. However, the mechanism by which metformin reduces diabetic asthma is yet to be clarified. This study aimed at ascertaining the downstream molecules underlying the effect of metformin on the activation of mast cells (MCs) and airway reactivity in a concomitant diabetic and asthmatic rat model. METHODS A T2DM model was induced utilizing a high-fat diet and streptozotocin. Then, 10% ovalbumin was utilized to stimulate asthma-like pathology in the T2DM rats. RBL-2H3 cells were induced by anti-dinitrophenyl-specific immunoglobulin E for constructing an in vitro model. Luciferase assay and RNA immunoprecipitation (IP) assay were conducted to identify the interaction between microRNA-152-3p (miR-152-3p) and DNA methyltransferase 1 (DNMT1), while chromatin IP to identify the binding of DNMT1 to insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF-1R) promoters. The effects of metformin on both pathological changes in vivo and biological behaviors of cells were evaluated. Using gain- and loss-of-function approaches, we assessed the role of the two interactions in the metformin-induced effect. RESULTS It was suggested that metformin could impede the MC activation and airway resistance in the concomitant diabetic and asthmatic rats. Additionally, metformin downregulated IR and IGF-1R through DNMT1-dependent methylation to repress MC activation and airway resistance. DNMT1 was testified to be a target gene of miR-152-3p. Furthermore, miR-152-3p-induced silencing of DNMT1 was blocked by metformin, hence restraining MC activation and airway resistance. CONCLUSION The findings cumulatively demonstrate that metformin downregulates IR/IGF-1R to block MC activation and airway resistance via impairing the binding affinity between miR-152-3p and DNMT1.
Collapse
Affiliation(s)
- Dan Fu
- Department of Endocrinology, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Hailu Zhao
- Diabetic Systems Center, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi, 541000, People's Republic of China
| | - Yan Huang
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Jingjuan Li
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Huafeng Feng
- Department of Anesthesiology, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People's Republic of China
| | - Aiguo Li
- Department of Anesthesiology, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People's Republic of China
| | - Yefen Liu
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China
| | - Liang He
- Department of Anesthesiology, The Fifth Affiliated Hospital of Southern Medical University, No.566, Congcheng Ave, Guangzhou, Guangdong, 510900, People's Republic of China.
| |
Collapse
|
16
|
Becker E, Husain M, Bone N, Smith S, Morris P, Zmijewski JW. AMPK activation improves recovery from pneumonia-induced lung injury via reduction of er-stress and apoptosis in alveolar epithelial cells. Respir Res 2023; 24:185. [PMID: 37438806 PMCID: PMC10337128 DOI: 10.1186/s12931-023-02483-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/24/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Bacterial pneumonia and related lung injury are among the most frequent causes of mortality in intensive care units, but also inflict serious and prolonged respiratory complications among survivors. Given that endoplasmic reticulum (ER) stress is a hallmark of sepsis-related alveolar epithelial cell (AEC) dysfunction, we tested if AMP-activated protein kinase (AMPK) affects recovery from ER stress and apoptosis of AECs during post-bacterial infection. METHODS In a murine model of lung injury by P. aeruginosa non-lethal infection, therapeutic interventions included AMPK activator metformin or GSK-3β inhibitor Tideglusib for 96 h. Recovery from AEC injury was evidenced by accumulation of soluble T-1α (AEC Type 1 marker) in BAL fluids along with fluorescence analysis of ER-stress (CHOP) and apoptosis (TUNEL) in lung sections. AMPK phosphorylation status and mediators of ER stress were determined via Immunoblot analysis from lung homogenates. Macrophage-dependent clearance of apoptotic cells was determined using flow cytometry assay. RESULTS P. aeruginosa-induced lung injury resulted in accumulation of neutrophils and cellular debris in the alveolar space along with persistent (96 h) ER-stress and apoptosis of AECs. While lung infection triggered AMPK inactivation (de-phosphorylation of Thr172-AMPK), metformin and Tideglusib promptly restored the AMPK activation status. In post infected mice, AMPK activation reduced indices of lung injury, ER stress and related apoptosis of AECs, as early as 24 h post administration of AMPK activators. In addition, we demonstrate that the extent of apoptotic cell accumulation is also dependent on AMPK-mediated clearance of apoptotic cells by macrophages. CONCLUSIONS Our study provides important insights into AMPK function in the preservation of AEC viability after bacterial infection, in particular due reduction of ER-stress and apoptosis, thereby promoting effective recovery from lung injury after pneumonia.
Collapse
Affiliation(s)
- Eugene Becker
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, 901 19th St. South BMRII 406, Birmingham, AL, 35294-0012, USA
| | - Maroof Husain
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, 901 19th St. South BMRII 406, Birmingham, AL, 35294-0012, USA
| | - Nathaniel Bone
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, 901 19th St. South BMRII 406, Birmingham, AL, 35294-0012, USA
| | - Samuel Smith
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, 901 19th St. South BMRII 406, Birmingham, AL, 35294-0012, USA
| | - Peter Morris
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, 901 19th St. South BMRII 406, Birmingham, AL, 35294-0012, USA
| | - Jaroslaw W Zmijewski
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, 901 19th St. South BMRII 406, Birmingham, AL, 35294-0012, USA.
| |
Collapse
|
17
|
Zhang R, Liang H, Liu G, Jiang W, Tang Z, Fan Q, Nie Z, Hu H, Kang G, Xie S. Nesfatin-1, a novel energy-regulating peptide, alleviates pulmonary fibrosis by blocking TGF-β1/Smad pathway in an AMPKα-dependent manner. Int Immunopharmacol 2023; 120:110369. [PMID: 37224650 DOI: 10.1016/j.intimp.2023.110369] [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: 02/27/2023] [Revised: 05/14/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023]
Abstract
Pulmonary fibrosis is a chronic progressive disease which steadily causes a critical public health concern. Nesfatin-1, a novel energy-regulating peptide discovered in 2006, could increase the level of AMPK phosphorylation. Previous studies have unveiled that Nesfatin-1 possessed many pharmacological effects including anti-inflammation, anti-oxidative stress, anti-fibrosis, and the regulation of lipid metabolism. Here, we investigated the impact of Nesfatin-1 on pulmonary fibrosis. Male C57BL/6J mice were intraperitoneally injected with Nesfatin-1 (10 μg·kg-1·day-1) for 21 days since mice were intratracheally administrated with bleomycin (BLM) (2 U/kg). Primary murine lung fibroblasts were stimulated with TGF-β1 (10 ng/ml) for 48 h. The results showed that Nesfatin-1 treatment significantly improved pulmonary function and decreased the production of collagen in BLM-treated mice. Meantime, Nesfatin-1 treatment also inhibited oxidative stress and inflammation in lung tissues from BLM-treated mice. Mechanically, Nesfatin-1 blocked the activation of TGF-β1/Smad2/3 signaling pathway in lung tissues challenged with BLM. In addition, we found that Nesfatin-1 enhanced the phosphorylation of AMPKα during pulmonary fibrosis. However, pharmacological inhibition or genetic deletion of AMPKα could both offset the pulmonary protection mediated by Nesfatin-1 during pulmonary fibrosis. Our experimental results firstly show Nesfatin-1 might serve as a novel treatment or adjuvant against pulmonary fibrosis by blocking TGF-β1/Smad pathway in an AMPKα-dependent manner.
Collapse
Affiliation(s)
- Renquan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hui Liang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Gaoli Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zheng Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qinglu Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhihao Nie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Haifeng Hu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ganjun Kang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Guerrero SC, Panettieri RA, Rastogi D. Mechanistic Links Between Obesity and Airway Pathobiology Inform Therapies for Obesity-Related Asthma. Paediatr Drugs 2023; 25:283-299. [PMID: 36656428 PMCID: PMC11071627 DOI: 10.1007/s40272-022-00554-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2022] [Indexed: 01/20/2023]
Abstract
Obesity-related asthma is associated with a high disease burden and a poor response to existent asthma therapies, suggesting that it is a distinct asthma phenotype. The proposed mechanisms that contribute to obesity-related asthma include the effects of the mechanical load of obesity, adipokine perturbations, and immune dysregulation. Each of these influences airway smooth muscle function. Mechanical fat load alters airway smooth muscle stretch affecting airway wall geometry, airway smooth muscle contractility, and agonist delivery; weight loss strategies, including medically induced weight loss, counter these effects. Among the metabolic disturbances, insulin resistance and free fatty acid receptor activation influence distinct signaling pathways in the airway smooth muscle downstream of both the M2 muscarinic receptor and the β2 adrenergic receptor, such as phospholipase C and the extracellular signal-regulated kinase signaling cascade. Medications that decrease insulin resistance and dyslipidemia are associated with a lower asthma disease burden. Leptin resistance is best understood to modulate muscarinic receptors via the neural pathways but there are no specific therapies for leptin resistance. From the immune perspective, monocytes and T helper cells are involved in systemic pro-inflammatory profiles driven by obesity, notably associated with elevated levels of interleukin-6. Clinical trials on tocilizumab, an anti-interleukin antibody, are ongoing for obesity-related asthma. This armamentarium of therapies is distinct from standard asthma medications, and once investigated for its efficacy and safety among children, will serve as a novel therapeutic intervention for pediatric obesity-related asthma. Irrespective of the directionality of the association between asthma and obesity, airway-specific mechanistic studies are needed to identify additional novel therapeutic targets for obesity-related asthma.
Collapse
Affiliation(s)
- Silvia Cabrera Guerrero
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, 111 Michigan Ave NW, Washington, DC, 20010, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Deepa Rastogi
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, 111 Michigan Ave NW, Washington, DC, 20010, USA.
| |
Collapse
|
19
|
Abstract
Increasing evidence suggests that there is acceleration of lung ageing in chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), with the accumulation of senescent cells in the lung. Senescent cells fail to repair tissue damage and release an array of inflammatory proteins, known as the senescence-associated secretory phenotype, which drive further senescence and disease progression. This suggests that targeting cellular senescence with senotherapies may treat the underlying disease process in COPD and IPF and thus reduce disease progression and mortality. Several existing or future drugs may inhibit the development of cellular senescence which is driven by chronic oxidative stress (senostatics), including inhibitors of PI3K-mTOR signalling pathways, antagomirs of critical microRNAs and novel antioxidants. Other drugs (senolytics) selectively remove senescent cells by promoting apoptosis. Clinical studies with senotherapies are already underway in chronic lung diseases.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart & Lung Institute, Imperial College London, United Kingdom.
| |
Collapse
|
20
|
Guarnieri G, Iervolino M, Cavallone S, Unfer V, Vianello A. The "Asthma-Polycystic Ovary Overlap Syndrome" and the Therapeutic Role of Myo-Inositol. Int J Mol Sci 2023; 24:ijms24086959. [PMID: 37108123 PMCID: PMC10138395 DOI: 10.3390/ijms24086959] [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: 02/21/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Asthma is a heterogeneous inflammatory disease characterized by abnormalities in immune response. Due to the inherent complexity of the disease and the presence of comorbidities, asthma control is often difficult to obtain. In asthmatic patients, an increased prevalence of irregular menstrual cycles, infertility, obesity, and insulin resistance has been reported. Given that these conditions are also common in patients with polycystic ovary syndrome (PCOS), we propose the definition of "asthma-PCOS overlap syndrome" to indicate a medical condition which shares characteristics of both diseases. The aim of this review is to analyze the links between asthma and PCOS and evaluate the therapeutic role of myo-inositol, a natural compound currently utilized in patients with PCOS, in the management of asthma patients.
Collapse
Affiliation(s)
- Gabriella Guarnieri
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | | | | | - Vittorio Unfer
- Systems Biology Group Laboratory, 00163 Rome, Italy
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy
| | - Andrea Vianello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| |
Collapse
|
21
|
Zhao Y, Zhang X, Wang G, Wu H, Chen R, Zhang Y, Yang S, Liu L. LXA4 inhibits TGF-β1-induced airway smooth muscle cells proliferation and migration by suppressing the Smad/YAP pathway. Int Immunopharmacol 2023; 118:110144. [PMID: 37030120 DOI: 10.1016/j.intimp.2023.110144] [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: 01/26/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/10/2023]
Abstract
The aims of the present study were to examine the signaling mechanisms for transforming growth factor-β1 (TGF-β1)-induced rat airway smooth muscle cells (ASMCs) proliferation and migration and to determine the effect of lipoxin A4 (LXA4) on TGF-β1-induced rat ASMCs proliferation and migration and its underlying mechanisms. TGF-β1 upregulated transcriptional coactivator Yes-associated protein (YAP) expression by activating Smad2/3 and then upregulated cyclin D1, leading to rat ASMCs proliferation and migration. This effect was reversed after treatment with the TGF-β1 receptor inhibitor SB431542. YAP is a critical mediator of TGF-β1-induced ASMCs proliferation and migration. Knockdown of YAP disrupted the pro-airway remodeling function of TGF-β1. Preincubation of rat ASMCs with LXA4 blocked TGF-β1-induced activation of Smad2/3 and changed its downstream targets, YAP and cyclin D1, resulting in the inhibition of rat ASMCs proliferation and migration. Our study suggests that LXA4 suppresses Smad/YAP signaling to inhibit rat ASMCs proliferation and migration and therefore has potential value in the prevention and treatment of asthma by negatively modulating airway remodeling.
Collapse
Affiliation(s)
- Yali Zhao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Xiangli Zhang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Guizuo Wang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Hua Wu
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Ruilin Chen
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Yongqing Zhang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Shumei Yang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China.
| |
Collapse
|
22
|
Matsubayashi S, Ito S, Araya J, Kuwano K. Drugs against metabolic diseases as potential senotherapeutics for aging-related respiratory diseases. Front Endocrinol (Lausanne) 2023; 14:1079626. [PMID: 37077349 PMCID: PMC10106576 DOI: 10.3389/fendo.2023.1079626] [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: 10/25/2022] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Recent advances in aging research have provided novel insights for the development of senotherapy, which utilizes cellular senescence as a therapeutic target. Cellular senescence is involved in the pathogenesis of various chronic diseases, including metabolic and respiratory diseases. Senotherapy is a potential therapeutic strategy for aging-related pathologies. Senotherapy can be classified into senolytics (induce cell death in senescent cells) and senomorphics (ameliorate the adverse effects of senescent cells represented by the senescence-associated secretory phenotype). Although the precise mechanism has not been elucidated, various drugs against metabolic diseases may function as senotherapeutics, which has piqued the interest of the scientific community. Cellular senescence is involved in the pathogenesis of chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), which are aging-related respiratory diseases. Large-scale observational studies have reported that several drugs, such as metformin and statins, may ameliorate the progression of COPD and IPF. Recent studies have reported that drugs against metabolic diseases may exert a pharmacological effect on aging-related respiratory diseases that can be different from their original effect on metabolic diseases. However, high non-physiological concentrations are needed to determine the efficacy of these drugs under experimental conditions. Inhalation therapy may increase the local concentration of drugs in the lungs without exerting systemic adverse effects. Thus, the clinical application of drugs against metabolic diseases, especially through an inhalation treatment modality, can be a novel therapeutic approach for aging-related respiratory diseases. This review summarizes and discusses accumulating evidence on the mechanisms of aging, as well as on cellular senescence and senotherapeutics, including drugs against metabolic diseases. We propose a developmental strategy for a senotherapeutic approach for aging-related respiratory diseases with a special focus on COPD and IPF.
Collapse
|
23
|
Uppal P, Mohammed SA, Rajashekar S, Giri Ravindran S, Kakarla M, Ausaja Gambo M, Yousri Salama M, Haidar Ismail N, Tavalla P, Hamid P. Type 2 Diabetes Mellitus and Asthma: Pathomechanisms of Their Association and Clinical Implications. Cureus 2023; 15:e36047. [PMID: 37056543 PMCID: PMC10089620 DOI: 10.7759/cureus.36047] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/11/2023] [Indexed: 03/14/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) and asthma are chronic illnesses concomitantly present in a significant percentage of the population. Their comorbidity is associated with poor disease control and lower quality of life, thus imposing a substantial medical and economic burden worldwide. This review investigates the association between asthma and T2DM, in terms of pathogenesis, clinical outcomes, and therapeutic opportunities. Our review found an increased risk of asthma among diabetics, and vice versa. Having diabetes and poor glycemic control is associated with an increased rate of asthma exacerbations and increased mortality among those hospitalized for asthma exacerbations. The mechanisms postulated for the diabetes-asthma association include chronic low-grade inflammation, obesity, hyperinsulinemia, and possibly diabetic pneumopathy. Usage of metformin, which is the first-line drug for type 2 diabetes, was found to be associated with a decreased asthma occurrence, asthma exacerbations, and asthma-related hospitalizations. Glucagon-like peptide 1 receptor agonists were also found to be associated with a lower occurrence of asthma exacerbations. Thiazolidinediones are also associated with lower rates of asthma exacerbations, but their clinical efficacy for the same was suggested to be limited. This literature review supports a partly causative association between asthma and diabetes. This comorbidity leads to poor patient compliance, worse disease outcomes, and poor quality of life. Thus, further studies are warranted to explore the prognostic implications, therapeutic opportunities, and specific clinical practice algorithms for patients with concurrent asthma and type 2 diabetes mellitus.
Collapse
|
24
|
Roshan Lal T, Cechinel LR, Freishtat R, Rastogi D. Metabolic Contributions to Pathobiology of Asthma. Metabolites 2023; 13:212. [PMID: 36837831 PMCID: PMC9962059 DOI: 10.3390/metabo13020212] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/11/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Asthma is a heterogenous disorder driven by inflammatory mechanisms that result in multiple phenotypes. Given the complex nature of this condition, metabolomics is being used to delineate the pathobiology of asthma. Metabolomics is the study of metabolites in biology, which includes biofluids, cells, and tissues. These metabolites have a vital role in a disease as they contribute to the pathogenesis of said condition. This review describes how macrometabolic and micrometabolic studies pertaining to these metabolites have contributed to our current understanding of asthma, as well as its many phenotypes. One of the main phenotypes this review will discuss in further detail is obesity as well as diabetes. Distinct roles of metabolites in endotyping asthma and their translation to potential therapy development for asthma is also discussed in this review.
Collapse
Affiliation(s)
- Tamanna Roshan Lal
- Rare Disease Institute, Children’s National Hospital, Washington, DC 20012, USA
| | - Laura Reck Cechinel
- Departments of Pediatrics and Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Robert Freishtat
- Departments of Pediatrics and Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Deepa Rastogi
- Departments of Pediatrics and Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| |
Collapse
|
25
|
Figueira-Gonçalves JM, Golpe R. Impact of Oral Antidiabetics Agents in the Prevention of COPD Exacerbations. Arch Bronconeumol 2022:S0300-2896(22)00667-6. [PMID: 36609104 DOI: 10.1016/j.arbres.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Juan Marco Figueira-Gonçalves
- Pneumology and Thoracic Surgery Service, Unit for Patients with Highly Complex COPD, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; University Institute of Tropical Disease and Public Health of the Canary Islands, University of La Laguna, Santa Cruz de Tenerife, Spain.
| | - Rafael Golpe
- Pneumology Service, University Hospital Lucus Augusti, Lugo, Spain
| |
Collapse
|
26
|
Kaplan AG, Kim JW. Asthma Exacerbations and Glucagon-Like Peptide-1 Receptor Agonists: a Review of the Current Evidence. Pulm Ther 2022; 8:343-358. [DOI: 10.1007/s41030-022-00203-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
|
27
|
Yang S, Wang L, Pan X, Liang Y, Zhang Y, Li J, Zhou B. 5-Methoxyflavone-induced AMPKα activation inhibits NF-κB and P38 MAPK signaling to attenuate influenza A virus-mediated inflammation and lung injury in vitro and in vivo. Cell Mol Biol Lett 2022; 27:82. [PMID: 36180831 PMCID: PMC9524045 DOI: 10.1186/s11658-022-00381-1] [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: 07/17/2022] [Accepted: 09/02/2022] [Indexed: 11/23/2022] Open
Abstract
Influenza-related acute lung injury (ALI) is a life-threatening condition that results mostly from uncontrolled replication of influenza virus (IV) and severe proinflammatory responses. The methoxy flavonoid compound 5-methoxyflavone (5-MF) is believed to have superior biological activity in the treatment of cancer. However, the effects and underlying mechanism of 5-MF on IV-mediated ALI are still unclear. Here, we showed that 5-MF significantly improved the survival of mice with lethal IV infection and ameliorated IV-mediated lung edema, lung histological changes, and inflammatory cell lung recruitment. We found that 5-MF has antiviral activity against influenza A virus (IAV), which was probably associated with increased expression of radical S-adenosyl methionine domain containing 2 (RSAD2) and suppression of endosomal acidification. Moreover, IV-infected A549 cells with 5-MF treatment markedly reduced proinflammatory mediator expression (IL-6, CXCL8, TNF-α, CXCL10, CCL2, CCL3, CCL4, GM-CSF, COX-2, and PGE2) and prevented P-IKBα, P-P65, and P-P38 activation. Interestingly, we demonstrated that 5-MF treatment could trigger activation of AMP-activated protein kinase (AMPK)α in IV-infected A549 cells, as evidenced by activation of the AMPKα downstream molecule P53. Importantly, the addition of AMPKα blocker compound C dramatically abolished 5-MF-mediated increased levels of RSAD2, the inhibitory effects on H1N1 virus-elicited endosomal acidification, and the suppression expression of proinflammatory mediators (IL-6, TNF-α, CXCL10, COX-2 and PGE2), as well as the inactivation of P-IKBα, P-P65, and P-P38 MAPK signaling pathways. Furthermore, inhibition of AMPKα abrogated the protective effects of 5-MF on H1N1 virus-mediated lung injury and excessive inflammation in vivo. Taken together, these results indicate that 5-MF alleviated IV-mediated ALI and suppressed excessive inflammatory responses through activation of AMPKα signaling.
Collapse
Affiliation(s)
- Sushan Yang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | | | | | - Yueyun Liang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Yuehan Zhang
- The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China. .,Institute of Chinese Integrative Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Beixian Zhou
- The People's Hospital of Gaozhou, Gaozhou, 525200, China.
| |
Collapse
|
28
|
Liang Z, Yang M, Xu C, Zeng R, Dong L. Effects and safety of metformin in patients with concurrent diabetes mellitus and chronic obstructive pulmonary disease: a systematic review and meta-analysis. Endocr Connect 2022; 11:e220289. [PMID: 35900801 PMCID: PMC9422254 DOI: 10.1530/ec-22-0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/21/2022] [Indexed: 11/15/2022]
Abstract
Aim This study aimed to investigate the effects and safety of metformin in patients with concurrent diabetes mellitus (DM) and chronic obstructive pulmonary disease (COPD). Methods PubMed, Embase, Web of Science, the China National Knowledge, and Cochrane Database were searched to find studies that examined the effects and safety of metformin in patients with concurrent DM and COPD. We conducted a meta-analysis with a risk ratio (RR) and assessed the quality of included studies and pooled evidence. Results Eight studies were involved. Metformin was associated with lower risk of COPD-related hospitalizations (RR: 0.72, 95% CI: 0.53-0.98; I2= 89%) and all-cause mortality (RR: 0.60, 95% CI: 0.36-1.01, I2= 69%) in patients with concurrent DM and COPD, but did not increase the risk of hyperlactatemia (RR: 1.14, 95% CI: 0.92-1.41, I2 = 8%). Conclusions Metformin use is associated with lower risk of COPD-related hospitalizations and risk of all-cause mortality without increasing the risk of hyperlactatemia. Considerations should be given to conduct more high-quality randomized trials involving larger samples.
Collapse
Affiliation(s)
- Ziting Liang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, The First Affiliated Hospital of Shandong First Medical University, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Mengge Yang
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, The First Affiliated Hospital of Shandong First Medical University, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Changjuan Xu
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, The First Affiliated Hospital of Shandong First Medical University, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Rong Zeng
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, The First Affiliated Hospital of Shandong First Medical University, Shandong Institute of Respiratory Diseases, Jinan, China
| | - Liang Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, The First Affiliated Hospital of Shandong First Medical University, Shandong Institute of Respiratory Diseases, Jinan, China
| |
Collapse
|
29
|
Zhang Y, Zhang H, Li S, Huang K, Jiang L, Wang Y. Metformin Alleviates LPS-Induced Acute Lung Injury by Regulating the SIRT1/NF-κB/NLRP3 Pathway and Inhibiting Endothelial Cell Pyroptosis. Front Pharmacol 2022; 13:801337. [PMID: 35910360 PMCID: PMC9334876 DOI: 10.3389/fphar.2022.801337] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS), a devastating complication of numerous conditions, is often associated with high mortality. It is well known that endothelial cell (EC) damage and inflammation are vital processes in the pathogenesis of ARDS. Nevertheless, the mechanisms of EC damage are largely unknown. In the present study, we investigated the role of pyroptosis in the initiation of ARDS and demonstrated that endothelial pyroptosis might play a pivotal role in the pathophysiology of ARDS. Metformin, an antidiabetic drug, exhibited a protective effect in lipopolysaccharide (LPS)-induced lung injury, and we hypothesized that metformin alleviated LPS-induced lung injury via inhibiting ECs pyroptosis. In vivo, male ICR mice were intratracheally injected with LPS, and metformin was previously administered intraperitoneally. Morphological properties of lung tissues were detected. We showed that metformin inhibited NLRP3 inflammasome activation and NLRP3-stimulated pyroptosis induction, as shown by decreased levels of cleaved caspase-1, N-terminal fragment of GSDMD, and protein contents of IL-1β in lung tissues of mice exposed to LPS. LPS-induced expression of vascular adhesion molecules was also reduced after the treatment with metformin. In vitro, exposure of pulmonary ECs to LPS resulted in increased expression of NLRP3 and pyroptosis-associated indicators. By inhibiting the expression of NLRP3 with NLRP3 inhibitor MCC950, pyroptosis-related markers and vascular adhesion molecules were ameliorated. Moreover, metformin treatment significantly inhibited the NF-κB signaling pathway and increased the expression of sirtuin 1 (SIRT1) both in LPS-stimulated lung tissues and pulmonary ECs. Administration of the selective SIRT1 inhibitor nicotinamide significantly reversed the protective effect of metformin against endothelial pyroptosis and lung injury in LPS-treated ECs and LPS-induced acute lung injury (ALI). Thus, these findings demonstrated that metformin alleviated LPS-induced ALI by inhibiting NF-κB-NLRP3–mediated ECs pyroptosis, possibly by upregulating the expression of SIRT1.
Collapse
Affiliation(s)
| | | | | | | | - Lai Jiang
- *Correspondence: Yan Wang, ; Lai Jiang,
| | - Yan Wang
- *Correspondence: Yan Wang, ; Lai Jiang,
| |
Collapse
|
30
|
Ramanathan R, Firdous A, Dong Q, Wang D, Lee J, Vo N, Sowa G. Investigation into the anti-inflammatory properties of metformin in intervertebral disc cells. JOR Spine 2022; 5:e1197. [PMID: 35783910 PMCID: PMC9238278 DOI: 10.1002/jsp2.1197] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Intervertebral disc degeneration (IDD) is closely related to heightened inflammation in the annulus fibrosis (AF) and nucleus pulposus (NP) cells in the intervertebral disc. An imbalanced matrix homeostasis has been shown to contribute to disc degeneration and associated discogenic low back pain. Metformin, a diabetes medication, has been noted to exhibit anti-inflammatory properties through upregulation of the AMPK pathway, leading to various anti-inflammatory-related responses in hepatocytes. However, it is still unclear how metformin influences disc cellular response to inflammatory stress and the corresponding mechanism. Hence, the objective of this study is to elucidate the effects of metformin on expression of key pro-inflammatory, catabolic, and anabolic factors within rat AF cells in response to inflammatory stimulation and mechanical tensile stress. Methods Five Fischer 344 rats were sacrificed and their spines isolated. AF cells were cultured and plated in flexible silicone membrane-based six-well plates. Wells were split into eight groups and subjected to metformin, IL-1β, mechanical stretch, and combined treatments. Relative gene expressions of MMP-13, COX-2, iNOS, AGC, and Col1 were assessed with quantitative real-time polymerase chain reaction (qRT-PCR), and downstream prostaglandin E2 (PGE2) production was quantified with enzyme-linked immunosorbent assay (ELISA). NF-kB nuclear translocation was also quantified. Results Metformin in the presence of the combined stress treatments (M + IL/S) significantly increased Col1, COX-2, and MMP-13 gene expression, decreased PGE2 production compared to IL/S conditions alone. Metformin treatment of cultured rat annulus fibrosus cells significantly reduced the nuclear translocation of NF-κB after 4 h of IL-1β treatment from 43.1% in case of IL-1β treatment down to 26.2% in the case of metformin + IL-1β treatment. Discussion The lack of metformin-mediated suppression of inflammatory response in the nonstretch groups indicates that metformin may be enacting its effects through a stretch-dependent manner. These results suggest a foundation for pursuing further research into metformin's potential role as an anti-inflammatory agent for curtailing intervertebral disc degeneration.
Collapse
Affiliation(s)
- Rahul Ramanathan
- Ferguson Spine Laboratory, Department of Orthopaedic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ayesha Firdous
- Ferguson Spine Laboratory, Department of Orthopaedic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Qing Dong
- Ferguson Spine Laboratory, Department of Orthopaedic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dong Wang
- Ferguson Spine Laboratory, Department of Orthopaedic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Joon Lee
- Ferguson Spine Laboratory, Department of Orthopaedic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Nam Vo
- Ferguson Spine Laboratory, Department of Orthopaedic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Gwendolyn Sowa
- Ferguson Spine Laboratory, Department of Orthopaedic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Physical Medicine and RehabilitationUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
31
|
Saunders RM, Biddle M, Amrani Y, Brightling CE. Stressed out - The role of oxidative stress in airway smooth muscle dysfunction in asthma and COPD. Free Radic Biol Med 2022; 185:97-119. [PMID: 35472411 DOI: 10.1016/j.freeradbiomed.2022.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/06/2022] [Accepted: 04/19/2022] [Indexed: 12/14/2022]
Abstract
The airway smooth muscle (ASM) surrounding the airways is dysfunctional in both asthma and chronic obstructive pulmonary disease (COPD), exhibiting; increased contraction, increased mass, increased inflammatory mediator release and decreased corticosteroid responsiveness. Due to this dysfunction, ASM is a key contributor to symptoms in patients that remain symptomatic despite optimal provision of currently available treatments. There is a significant body of research investigating the effects of oxidative stress/ROS on ASM behaviour, falling into the following categories; cigarette smoke and associated compounds, air pollutants, aero-allergens, asthma and COPD relevant mediators, and the anti-oxidant Nrf2/HO-1 signalling pathway. However, despite a number of recent reviews addressing the role of oxidative stress/ROS in asthma and COPD, the potential contribution of oxidative stress/ROS-related ASM dysfunction to asthma and COPD pathophysiology has not been comprehensively reviewed. We provide a thorough review of studies that have used primary airway, bronchial or tracheal smooth muscle cells to investigate the role of oxidative stress/ROS in ASM dysfunction and consider how they could contribute to the pathophysiology of asthma and COPD. We summarise the current state of play with regards to clinical trials/development of agents targeting oxidative stress and associated limitations, and the adverse effects of oxidative stress on the efficacy of current therapies, with reference to ASM related studies where appropriate. We also identify limitations in the current knowledge of the role of oxidative stress/ROS in ASM dysfunction and identify areas for future research.
Collapse
Affiliation(s)
- Ruth M Saunders
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK.
| | - Michael Biddle
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Yassine Amrani
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Christopher E Brightling
- The Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
32
|
Kazmi S, Khan MA, Shamma T, Altuhami A, Assiri AM, Broering DC. Therapeutic nexus of T cell immunometabolism in improving transplantation immunotherapy. Int Immunopharmacol 2022; 106:108621. [DOI: 10.1016/j.intimp.2022.108621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/03/2022] [Accepted: 02/10/2022] [Indexed: 11/26/2022]
|
33
|
Gu C, Loube J, Lee R, Bevans-Fonti S, Wu TD, Barmine JH, Jun JC, McCormack MC, Hansel NN, Mitzner W, Polotsky VY. Metformin Alleviates Airway Hyperresponsiveness in a Mouse Model of Diet-Induced Obesity. Front Physiol 2022; 13:883275. [PMID: 35574481 PMCID: PMC9098833 DOI: 10.3389/fphys.2022.883275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/14/2022] [Indexed: 12/03/2022] Open
Abstract
Obese asthma is a unique phenotype of asthma characterized by non-allergic airway hyperresponsiveness (AHR) and inflammation which responds poorly to standard asthma therapy. Metformin is an oral hypoglycemic drug with insulin-sensitizing and anti-inflammatory properties. The objective of the current study was to test the effect of metformin on AHR in a mouse model of diet-induced obesity (DIO). We fed 12-week-old C57BL/6J DIO mice with a high fat diet for 8 weeks and treated them with either placebo (control, n = 10) or metformin (n = 10) added in drinking water (300 mg/kg/day) during the last 2 weeks of the experiment. We assessed AHR, metabolic profiles, and inflammatory markers after treatments. Metformin did not affect body weight or fasting blood glucose, but significantly reduced serum insulin (p = 0.0117). Metformin reduced AHR at 30 mg/ml of methacholine challenge (p = 0.0052) without affecting baseline airway resistance. Metformin did not affect circulating white blood cell counts or lung cytokine mRNA expression, but modestly decreased circulating platelet count. We conclude that metformin alleviated AHR in DIO mice. This finding suggests metformin has the potential to become an adjuvant pharmacological therapy in obese asthma.
Collapse
Affiliation(s)
- Chenjuan Gu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jeff Loube
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Rachel Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shannon Bevans-Fonti
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Tianshi David Wu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine and the Center for Innovations in Quality, Effectiveness, and Safety, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| | - Jessica H. Barmine
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan C. Jun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Meredith C. McCormack
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nadia N. Hansel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Vsevolod Y. Polotsky,
| |
Collapse
|
34
|
Teague TT, Payne SR, Kelly BT, Dempsey TM, McCoy RG, Sangaralingham LR, Limper AH. Evaluation for clinical benefit of metformin in patients with idiopathic pulmonary fibrosis and type 2 diabetes mellitus: a national claims-based cohort analysis. Respir Res 2022; 23:91. [PMID: 35410255 PMCID: PMC9004115 DOI: 10.1186/s12931-022-02001-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/21/2022] [Indexed: 12/27/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with high morbidity and limited treatment options. Type 2 diabetes mellitus (T2DM) is a common comorbid illness among patients with IPF and is often treated with metformin, the first-line agent in the management of T2DM. There is growing evidence demonstrating metformin’s anti-fibrotic properties; however, there is little real-world clinical data regarding its potential effectiveness in IPF. This study aims to evaluate the clinical benefit of metformin in patients with IPF and T2DM. Methods This nationwide cohort study used de-identified administrative claims data from OptumLabs® Data Warehouse to identify 3599 adults with IPF and concomitant T2DM between January 1, 2014 and June 30, 2019. Two cohorts were created: a cohort treated with metformin (n = 1377) and a cohort not treated with metformin (n = 2222). A final 1:1 propensity score-matched cohort compared 1100 patients with IPF and T2DM receiving metformin to those with both diagnoses but not receiving metformin; matching accounted for age, sex, race/ethnicity, residence region, year, medications, oxygen use, smoking status, healthcare use, and comorbidities. Outcomes were all-cause mortality (primary) and hospitalizations (secondary). Results Among 2200 patients with IPF and T2DM included in this matched analysis, metformin therapy was associated with a reduction in all-cause mortality (hazard ratio [HR], 0.46; 95% confidence interval [CI], 0.36–0.58; p < 0.001) and hospitalizations (HR, 0.82; 95% CI, 0.72–0.93; p = 0.003) compared to patients not receiving metformin. Conclusions Among patients with IPF and T2DM, metformin therapy may be associated with improved clinical outcomes. However, further investigation with randomized clinical trials is necessary prior to metformin’s broad implementation in the clinical management of IPF.
Collapse
|
35
|
Hollenberg MD, Epstein M. The innate immune response, microenvironment proteinases, and the COVID-19 pandemic: pathophysiologic mechanisms and emerging therapeutic targets. Kidney Int Suppl (2011) 2022; 12:48-62. [PMID: 35316977 PMCID: PMC8931295 DOI: 10.1016/j.kisu.2021.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/19/2021] [Accepted: 12/11/2021] [Indexed: 12/13/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, causing considerable mortality and morbidity worldwide, has fully engaged the biomedical community in attempts to elucidate the pathophysiology of COVID-19 and develop robust therapeutic strategies. To this end, the predominant research focus has been on the adaptive immune response to COVID-19 infections stimulated by mRNA and protein vaccines and on the duration and persistence of immune protection. In contrast, the role of the innate immune response to the viral challenge has been underrepresented. This overview focuses on the innate immune response to COVID-19 infection, with an emphasis on the roles of extracellular proteases in the tissue microenvironment. Proteinase-mediated signaling caused by enzymes in the extracellular microenvironment occurs upstream of the increased production of inflammatory cytokines that mediate COVID-19 pathology. These enzymes include the coagulation cascade, kinin-generating plasma kallikrein, and the complement system, as well as angiotensin-generating proteinases of the renin-angiotensin system. Furthermore, in the context of several articles in this Supplement elucidating and detailing the trajectory of diverse profibrotic pathways, we extrapolate these insights to explore how fibrosis and profibrotic pathways participate importantly in the pathogenesis of COVID-19. We propose that the lessons garnered from understanding the roles of microenvironment proteinases in triggering the innate immune response to COVID-19 pathology will identify potential therapeutic targets and inform approaches to the clinical management of COVID-19. Furthermore, the information may also provide a template for understanding the determinants of COVID-19-induced tissue fibrosis that may follow resolution of acute infection (so-called "long COVID"), which represents a major new challenge to our healthcare systems.
Collapse
Affiliation(s)
- Morley D. Hollenberg
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Murray Epstein
- Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
36
|
Tao F, Zhou Y, Wang M, Wang C, Zhu W, Han Z, Sun N, Wang D. Metformin alleviates chronic obstructive pulmonary disease and cigarette smoke extract-induced glucocorticoid resistance by activating the nuclear factor E2-related factor 2/heme oxygenase-1 signaling pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:95-111. [PMID: 35203060 PMCID: PMC8890943 DOI: 10.4196/kjpp.2022.26.2.95] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/08/2021] [Accepted: 11/29/2021] [Indexed: 11/15/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is an important healthcare problem worldwide. Often, glucocorticoid (GC) resistance develops during COPD treatment. As a classic hypoglycemic drug, metformin (MET) can be used as a treatment strategy for COPD due to its anti-inflammatory and antioxidant effects, but its specific mechanism of action is not known. We aimed to clarify the role of MET on COPD and cigarette smoke extract (CSE)-induced GC resistance. Through establishment of a COPD model in rats, we found that MET could improve lung function, reduce pathological injury, as well as reduce the level of inflammation and oxidative stress in COPD, and upregulate expression of nuclear factor E2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), multidrug resistance protein 1 (MRP1), and histone deacetylase 2 (HDAC2). By establishing a model of GC resistance in human bronchial epithelial cells stimulated by CSE, we found that MET reduced secretion of interleukin-8, and could upregulate expression of Nrf2, HO-1, MRP1, and HDAC2. MET could also increase the inhibition of MRP1 efflux by MK571 significantly, and increase expression of HDAC2 mRNA and protein. In conclusion, MET may upregulate MRP1 expression by activating the Nrf2/HO-1 signaling pathway, and then regulate expression of HDAC2 protein to reduce GC resistance.
Collapse
Affiliation(s)
- Fulin Tao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Yuanyuan Zhou
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Mengwen Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Chongyang Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Wentao Zhu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Zhili Han
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Nianxia Sun
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Dianlei Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, Anhui 230012, China
| |
Collapse
|
37
|
Huang M, Jiang W, Luo C, Yang M, Ren Y. Atractylenolide III inhibits epithelial‑mesenchymal transition in small intestine epithelial cells by activating the AMPK signaling pathway. Mol Med Rep 2022; 25:98. [PMID: 35088892 PMCID: PMC8809054 DOI: 10.3892/mmr.2022.12614] [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: 06/08/2021] [Accepted: 09/17/2021] [Indexed: 11/28/2022] Open
Abstract
Compared with the available drugs for the treatment of fibrosis in other organs, the development of intestinal anti-fibrosis drugs is limited. Therefore, it is of practical significance to examine novel drugs to delay or block the development of intestinal fibrosis. The present study aimed to investigate the effect of atractylenolide III (ATL-III) on intestinal fibrosis. An MTT assay was used to detect the effect of ATL-III on the activity of IEC-6 cells. The migration and invasion of fibrotic cells stimulated with TGF-β were determined via wound healing and Transwell assays. An immunofluorescence assay and western blotting were conducted to assess the expression levels of protein associated with epithelial-mesenchymal transition (EMT). The role of the AMP-activated protein kinase (AMPK) pathway was verified using compound C (an AMPK inhibitor) treatment. The results of the present study indicated that ATL-III had no effect on the cells at a dose of 1–20 µmol/l. Moreover, ATL-III can inhibit the invasion and migration of cells induced by TGF-β1, as well as block the EMT process. It was found that ATL-III could also activate the AMPK pathway. Furthermore, compound C reduced the inhibitory effect of ATL-III on stimulated cells, which indicated that the AMPK pathway plays a role in the inhibition process. In conclusion, ATL-III may inhibit the EMT of IEC-6 cells stimulated with TGF-β1 by activating the AMPK signaling pathway.
Collapse
Affiliation(s)
- Mingjin Huang
- College of Agriculture, Guizhou University, Guiyang, Guizhou 550025, P.R. China
| | - Wenwen Jiang
- College of Pharmaceutical Science, Guizhou University, Guiyang, Guizhou 550025, P.R. China
| | - Chunli Luo
- College of Agriculture, Guizhou University, Guiyang, Guizhou 550025, P.R. China
| | - Min Yang
- College of Pharmaceutical Science, Guizhou University, Guiyang, Guizhou 550025, P.R. China
| | - Yan Ren
- College of Pharmaceutical Science, Guizhou University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
38
|
Ma W, Jin Q, Guo H, Han X, Xu L, Lu S, Wu C. Metformin Ameliorates Inflammation and Airway Remodeling of Experimental Allergic Asthma in Mice by Restoring AMPKα Activity. Front Pharmacol 2022; 13:780148. [PMID: 35153777 PMCID: PMC8830934 DOI: 10.3389/fphar.2022.780148] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/03/2022] [Indexed: 12/19/2022] Open
Abstract
Metformin has been involved in modulating inflammatory state and inhibiting cell proliferation and angiogenesis. This study aimed to determine whether metformin alleviates airway inflammation and remodeling of experimental allergic asthma and elucidate the underlying mechanism. We sensitized and challenged mice with ovalbumin (OVA) to induce allergic asthma. During the challenge period, metformin was administered by intraperitoneal injection. By histopathological and immunohistochemical analyses, metformin-treated mice showed a significant alleviation in airway inflammation, and in the parameters of airway remodeling including goblet cell hyperplasia, collagen deposition and airway smooth muscle hypertrophy compared to those in the OVA-challenged mice. We also observed elevated levels of multiple cytokines (IL-4, IL-5, IL-13, TNF-α, TGF-β1 and MMP-9) in the bronchoalveolar lavage fluid, OVA-specific IgE in the serum and angiogenesis-related factors (VEGF, SDF-1 and CXCR4) in the plasma from asthmatic mice, while metformin reduced all these parameters. Additionally, the activity of 5′-adenosine monophosphate-activated protein kinase a (AMPKα) in the lungs from OVA-challenged mice was remarkably lower than control ones, while after metformin treatment, the ratio of p-AMPKα to AMPKα was upregulated and new blood vessels in the sub-epithelial area as evidenced by CD31 staining were effectively suppressed. These results indicate that metformin ameliorates airway inflammation and remodeling in an OVA-induced chronic asthmatic model and its protective role could be associated with the restoration of AMPKα activity and decreased asthma-related angiogenesis.
Collapse
Affiliation(s)
- Wenxian Ma
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Qiaoyan Jin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Haiqin Guo
- Department of Pulmonary and Critical Care Medicine, Third Military Medical University Southwest Hospital, Chongqing, China
| | - Xinpeng Han
- Department of Pulmonary and Critical Care Medicine, Xi’an International Medical Center Hospital, Xi’an, China
| | - Lingbin Xu
- Department of Pulmonary and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- *Correspondence: Changgui Wu, ; Shemin Lu,
| | - Changgui Wu
- Department of Pulmonary and Critical Care Medicine, Xi’an International Medical Center Hospital, Xi’an, China
- *Correspondence: Changgui Wu, ; Shemin Lu,
| |
Collapse
|
39
|
Ahmad I, Molyvdas A, Jian MY, Zhou T, Traylor AM, Cui H, Liu G, Song W, Agarwal A, Jilling T, Aggarwal S, Matalon S. AICAR decreases acute lung injury by phosphorylating AMPK and upregulating heme oxygenase-1. Eur Respir J 2021; 58:2003694. [PMID: 34049949 PMCID: PMC9144003 DOI: 10.1183/13993003.03694-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/11/2021] [Indexed: 11/05/2022]
Abstract
AIM We investigated the mechanisms by which N1-(β-d-ribofuranosyl)-5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an activator of AMP-activated protein kinase (AMPK), decreases lung injury and mortality when administered to mice post exposure to bromine gas (Br2). METHODS We exposed male C57BL/6 mice and heme oxygenase-1 (HO-1)-deficient (HO-1-/-) and corresponding wild-type (WT) littermate mice to Br2 (600 ppm for 45 or 30 min, respectively) in environmental chambers and returned them to room air. AICAR was administered 6 h post exposure (10 mg·kg-1, intraperitoneal). We assessed survival, indices of lung injury, high mobility group box 1 (HMGB1) in the plasma, HO-1 levels in lung tissues and phosphorylation of AMPK and its upstream liver kinase B1 (LKB1). Rat alveolar type II epithelial (L2) cells and human club-like epithelial (H441) cells were also exposed to Br2 (100 ppm for 10 min). After 24 h we measured apoptosis and necrosis, AMPK and LKB1 phosphorylation, and HO-1 expression. RESULTS There was a marked downregulation of phosphorylated AMPK and LKB1 in lung tissues and in L2 and H441 cells post exposure. AICAR increased survival in C57BL/6 but not in HO-1-/- mice. In WT mice, AICAR decreased lung injury and restored phosphorylated AMPK and phosphorylated LKB1 to control levels and increased HO-1 levels in both lung tissues and cells exposed to Br2. Treatment of L2 and H441 cells with small interfering RNAs against nuclear factor erythroid 2-related factor 2 or HO-1 abrogated the protective effects of AICAR. CONCLUSIONS Our data indicate that the primary mechanism for the protective action of AICAR in toxic gas injury is the upregulation of lung HO-1 levels.
Collapse
Affiliation(s)
- Israr Ahmad
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally to this study
| | - Adam Molyvdas
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally to this study
| | - Ming-Yuan Jian
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ting Zhou
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amie M Traylor
- Division of Nephrology, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Weifeng Song
- Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Division of Nephrology, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tamas Jilling
- Division of Neonatology, Dept of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally as senior authors
| | - Sadis Matalon
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally as senior authors
| |
Collapse
|
40
|
Blunting p38 MAPKα and ERK1/2 activities by empagliflozin enhances the antifibrotic effect of metformin and augments its AMPK-induced NF-κB inactivation in mice intoxicated with carbon tetrachloride. Life Sci 2021; 286:120070. [PMID: 34688695 DOI: 10.1016/j.lfs.2021.120070] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/09/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022]
Abstract
AIM Metformin and empagliflozin combined therapy may have complementary effects that go beyond the well-recognized targets of their monotherapy through AMPK activation. Therefore, the current study was designed to investigate for the first time the hepatoprotective effects of such combination therapy in the carbon tetrachloride (CCl4)-induced hepatic fibrosis model in mice. MATERIALS AND METHODS Determination of liver enzymes and the liver content of oxidative stress parameters, and hydroxyproline were performed biochemically. ELISA was performed to measure PDGF-BB, TNF-α, TGF-β, TIMP-1, AMPK, p-mTOR, NF-κB P65 binding activity, p38 MAPKα, JNK1/2 and ERK1/2. Real-time qPCR was conducted to determine Col1a1 and α-SMA. In addition, histopathological examination using H&E and Masson's trichrome stain were performed for determination of histopathological changes. KEY FINDINGS Empagliflozin inhibited the activation of p38 MAPK and ERK1/2 and exhibited a weak AMPKα stimulation. On the other hand, metformin exerted a more robust stimulatory action on the AMPKα that was accompanied by a notable decrease in the NF-κB nuclear binding activity and a decline in the p-mTOR levels. Nevertheless, the effect of metformin on MAPK kinases was insignificant. Our results revealed that blunting p38 MAPKα and ERK1/2 activities by empagliflozin enhanced the antifibrotic effect of metformin and augmented its AMPK-induced NF-κB inactivation. SIGNIFICANCE As diabetes is one of the most common risk factors for liver fibrosis, the use of antidiabetic drugs is expected to improve therapeutic outcome. Therefore, metformin/empagliflozin combined therapy could be promising in preventing hepatic inflammation and fibrosis via exhibiting complementary effects particularly in diabetic patients.
Collapse
|
41
|
Metformin effect in models of inflammation is associated with activation of ATP-dependent potassium channels and inhibition of tumor necrosis factor-α production. Inflammopharmacology 2021; 30:233-241. [PMID: 34850313 DOI: 10.1007/s10787-021-00899-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/21/2021] [Indexed: 10/19/2022]
Abstract
Metformin is an oral hypoglycemic drug widely used in the management of type 2 diabetes mellitus. We have recently demonstrated that metformin exhibits activity in models of nociceptive and neuropathic pain. However, little is known about its effects in experimental models of inflammation and inflammatory pain. Thus, the present study aimed to evaluate the activity of metformin in experimental models of inflammation and inflammatory pain in mice, as well as the underlying mechanisms. Previous (1 h) per os (p.o.) administration of metformin (250, 500 or 1000 mg/kg) inhibited the mechanical allodynia and paw edema induced by intraplantar (i.pl.) injection of carrageenan (600 μg) and also the pleurisy induced by this stimulus (200 μg, intrapleural). In the model of mechanical allodynia and paw edema induced by carrageenan, metformin also exhibited activity when administered after (1 h) the inflammatory stimulus. Metformin (1000 mg/kg) reduced the production of tumor necrosis factor-α induced by i.pl. injection of carrageenan. Metformin antiallodynic effect was not affected by previous administration of naltrexone (5 or 10 mg/kg, intraperitoneal) or cyproheptadine (5 or 10 mg/kg, p.o). However, this effect was abolished by previous administration of glibenclamide (20 or 40 mg/kg, p.o). In conclusion, the results demonstrate the activity of metformin in models of inflammation and inflammatory pain. In addition, the results indicate that the activity of metformin may be mediated by activation of ATP-sensitive potassium channels and reduction of production of inflammatory mediators. Altogether, these results stimulate the conduction of studies aiming to evaluate whether metformin may be repositioned in the treatment of patients with painful and inflammatory disorders.
Collapse
|
42
|
Therapeutic approaches targeting molecular signaling pathways common to diabetes, lung diseases and cancer. Adv Drug Deliv Rev 2021; 178:113918. [PMID: 34375681 DOI: 10.1016/j.addr.2021.113918] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus (DM), is the most common metabolic disease and is characterized by sustained hyperglycemia. Accumulating evidences supports a strong association between DM and numerous lung diseases including chronic obstructive pulmonary disease (COPD), fibrosis, and lung cancer (LC). The global incidence of DM-associated lung disorders is rising and several ongoing studies, including clinical trials, aim to elucidate the molecular mechanisms linking DM with lung disorders, in particular LC. Several potential mechanisms, including hyperglycemia, hyperinsulinemia, glycation, inflammation, and hypoxia, are cited as plausible links between DM and LC. In addition, studies also propose a connection between the use of anti-diabetic medications and reduction in the incidence of LC. However, the exact cause for DM associated lung diseases especially LC is not clear and is an area under intense investigation. Herein, we review the biological links reported between DM and lung disorders with an emphasis on LC. Furthermore, we report common signaling pathways (eg: TGF-β, IL-6, HIF-1, PDGF) and miRNAs that are dysregulated in DM and LC and serve as molecular targets for therapy. Finally, we propose a nanomedicine based approach for delivering therapeutics (eg: IL-24 plasmid DNA, HuR siRNA) to disrupt signaling pathways common to DM and LC and thus potentially treat DM-associated LC. Finally, we conclude that the effective modulation of commonly regulated signaling pathways would help design novel therapeutic protocols for treating DM patients diagnosed with LC.
Collapse
|
43
|
Schuliga M, Read J, Knight DA. Ageing mechanisms that contribute to tissue remodeling in lung disease. Ageing Res Rev 2021; 70:101405. [PMID: 34242806 DOI: 10.1016/j.arr.2021.101405] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/13/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Age is a major risk factor for chronic respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and certain phenotypes of asthma. The recent COVID-19 pandemic also highlights the increased susceptibility of the elderly to acute respiratory distress syndrome (ARDS), a diffuse inflammatory lung injury with often long-term effects (ie parenchymal fibrosis). Collectively, these lung conditions are characterized by a pathogenic reparative process that, rather than restoring organ function, contributes to structural and functional tissue decline. In the ageing lung, the homeostatic control of wound healing following challenge or injury has an increased likelihood of being perturbed, increasing susceptibility to disease. This loss of fidelity is a consequence of a diverse range of underlying ageing mechanisms including senescence, mitochondrial dysfunction, proteostatic stress and diminished autophagy that occur within the lung, as well as in other tissues, organs and systems of the body. These ageing pathways are highly interconnected, involving localized and systemic increases in inflammatory mediators and damage associated molecular patterns (DAMPs); along with corresponding changes in immune cell function, metabolism and composition of the pulmonary and gut microbiomes. Here we comprehensively review the roles of ageing mechanisms in the tissue remodeling of lung disease.
Collapse
Affiliation(s)
- Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| | - Jane Read
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
44
|
Li SX, Li C, Pang XR, Zhang J, Yu GC, Yeo AJ, Lavin MF, Shao H, Jia Q, Peng C. Metformin Attenuates Silica-Induced Pulmonary Fibrosis by Activating Autophagy via the AMPK-mTOR Signaling Pathway. Front Pharmacol 2021; 12:719589. [PMID: 34434111 PMCID: PMC8381252 DOI: 10.3389/fphar.2021.719589] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Long-term exposure to crystalline silica particles leads to silicosis characterized by persistent inflammation and progressive fibrosis in the lung. So far, there is no specific treatment to cure the disease other than supportive care. In this study, we examined the effects of metformin, a prescribed drug for type || diabetes on silicosis and explored the possible mechanisms in an established rat silicosis model in vivo, and an in vitro co-cultured model containing human macrophages cells (THP-1) and human bronchial epithelial cells (HBEC). Our results showed that metformin significantly alleviated the inflammation and fibrosis of lung tissues of rats exposed to silica particles. Metformin significantly reduced silica particle-induced inflammatory cytokines including transforming growth factor-β1 (TGF-β1), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in rat lung tissue and HBEC culture supernatant. The protein levels of Vimentin and α-smooth muscle actin (α-SMA) were significantly decreased by metfomin while expression level of E-cadherin (E-Cad) increased. Besides, metformin increased the expression levels of phosphorylated adenosine 5′-monophosphate (AMP)-activated protein kinase (p-AMPK), microtubule-associated protein (MAP) light chain 3B (LC3B) and Beclin1 proteins, and reduced levels of phosphorylated mammalian target of rapamycin (p-mTOR) and p62 proteins in vivo and in vitro. These results suggest that metformin could inhibit silica-induced pulmonary fibrosis by activating autophagy through the AMPK-mTOR pathway.
Collapse
Affiliation(s)
- Shu-Xian Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xin-Ru Pang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Juan Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Gong-Chang Yu
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Abrey J Yeo
- University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD, Australia
| | - Martin F Lavin
- University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD, Australia
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Cheng Peng
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Disorders of glucose metabolism, including insulin resistance, prediabetes, and diabetes, have been identified as risk factors for worsened asthma. This review summarizes emerging evidence for their role as modifiable risk factors in asthma, including the potential benefit of diabetes medications on asthma outcomes. RECENT FINDINGS Experimental studies show that hyperinsulinemia associated with insulin resistance is associated with airway smooth muscle proliferation and promotes contractility. Epidemiologic studies have identified a higher prevalence of glycemic dysfunction among those with severe and uncontrolled asthma, and longitudinal studies have associated prediabetes and diabetes with higher risk of asthma exacerbations. The potential benefits of thiazolidinediones (TZDs), glucagon-like peptide-1 agonists, and metformin being investigated in asthma, but thus far interventional studies of TZDs have reported null results. On the contrary, observational studies have inconsistently controlled for relevant confounders which leaves conclusions vulnerable to misattribution of relationships due to corelated metabolic disorders, including dyslipidemia. SUMMARY Developing evidence suggests that disorders of glucose metabolism may be associated with worsening asthma. However, these conditions arise within a network of obesity-related metabolic diseases that may themselves worsen asthma. Few interventional trials have not identified a benefit, but data have been limited. Additional research is needed to define the potential independent impact of disorders of glucose metabolism in asthma.
Collapse
|
46
|
Asgharzadeh F, Barneh F, Fakhraie M, Adel Barkhordar SL, Shabani M, Soleimani A, Rahmani F, Ariakia F, Mehraban S, Avan A, Hashemzehi M, Arjmand MH, Behnam-Rassouli R, Jaberi N, Sayyed-Hosseinian SH, Ferns GA, Ryzhikov M, Jafari M, Khazaei M, Hassanian SM. Metformin inhibits polyphosphate-induced hyper-permeability and inflammation. Int Immunopharmacol 2021; 99:107937. [PMID: 34271418 DOI: 10.1016/j.intimp.2021.107937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/09/2021] [Accepted: 06/28/2021] [Indexed: 12/19/2022]
Abstract
Circulating inflammatory factor inorganic polyphosphate (polyP) released from activated platelets could enhance factor XII and bradykinin resulted in increased capillary leakage and vascular permeability. PolyP induce inflammatory responses through mTOR pathway in endothelial cells, which is being reported in several diseases including atherosclerosis, thrombosis, sepsis, and cancer. Systems and molecular biology approaches were used to explore the regulatory role of the AMPK activator, metformin, on polyP-induced hyper-permeability in different organs in three different models of polyP-induced hyper-permeability including local, systemic short- and systemic long-term approaches in murine models. Our results showed that polyP disrupts endothelial barrier integrity in skin, liver, kidney, brain, heart, and lung in all three study models and metformin abrogates the disruptive effect of polyP. We also showed that activation of AMPK signaling pathway, regulation of oxidant/anti-oxidant balance, as well as decrease in inflammatory cell infiltration constitute a set of molecular mechanisms through which metformin elicits it's protective responses against polyP-induced hyper-permeability. These results support the clinical values of AMPK activators including the FDA-approved metformin in attenuating vascular damage in polyP-associated inflammatory diseases.
Collapse
Affiliation(s)
- Fereshteh Asgharzadeh
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farnaz Barneh
- Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Maryam Fakhraie
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohammad Shabani
- Department of Medical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atena Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Fatemeh Ariakia
- Department of Medical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saeedeh Mehraban
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Milad Hashemzehi
- Tropical and Communicable Diseases Research Centre, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Reyhaneh Behnam-Rassouli
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Najmeh Jaberi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, MO, USA
| | - Mohieddin Jafari
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
47
|
Bolívar S, Noriega L, Ortega S, Osorio E, Rosales W, Mendoza X, Mendoza-Torres E. Novel Targets of Metformin in Cardioprotection: Beyond the Effects Mediated by AMPK. Curr Pharm Des 2021; 27:80-90. [PMID: 32386485 DOI: 10.2174/1381612826666200509232610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/30/2020] [Indexed: 11/22/2022]
Abstract
Ischemic heart disease is the main cause of death globally. In the heart, the ischemia/reperfusion injury gives rise to a complex cascade of molecular signals, called cardiac remodeling, which generates harmful consequences for the contractile function of the myocardium and consequently heart failure. Metformin is the drug of choice in the treatment of type 2 diabetes mellitus. Clinical data suggest the direct effects of this drug on cardiac metabolism and studies in animal models showed that metformin activates the classical pathway of AMP-activated protein kinase (AMPK), generating cardioprotective effects during cardiac remodeling, hypertrophy and fibrosis. Furthermore, new studies have emerged about other targets of metformin with a potential role in cardioprotection. This state of the art review shows the available scientific evidence of the cardioprotective potential of metformin and its possible effects beyond AMPK. Targeting of autophagy, mitochondrial function and miRNAs are also explored as cardioprotective approaches along with a therapeutic potential. Further advances related to the biological effects of metformin and cardioprotective approaches may provide new therapies to protect the heart and prevent cardiac remodeling and heart failure.
Collapse
Affiliation(s)
- Samir Bolívar
- Healthcare Pharmacy and Pharmacology Research Group, Facultad de Quimica y Farmacia, Universidad del Atlantico, Barranquilla, Colombia
| | - Laura Noriega
- Healthcare Pharmacy and Pharmacology Research Group, Facultad de Quimica y Farmacia, Universidad del Atlantico, Barranquilla, Colombia
| | - Stefany Ortega
- Healthcare Pharmacy and Pharmacology Research Group, Facultad de Quimica y Farmacia, Universidad del Atlantico, Barranquilla, Colombia
| | - Estefanie Osorio
- Advaced Research Group in Biomedicine, Universidad Libre Seccional Barranquilla, Colombia
| | - Wendy Rosales
- Advaced Research Group in Biomedicine, Universidad Libre Seccional Barranquilla, Colombia
| | - Xilene Mendoza
- Universidad Metropolitana, Grupo de Investigacion en Medicina Traslacional (GIMET), Barranquilla, Colombia
| | - Evelyn Mendoza-Torres
- Advaced Research Group in Biomedicine, Universidad Libre Seccional Barranquilla, Colombia
| |
Collapse
|
48
|
Ma B, Athari SS, Mehrabi Nasab E, Zhao L. PI3K/AKT/mTOR and TLR4/MyD88/NF-κB Signaling Inhibitors Attenuate Pathological Mechanisms of Allergic Asthma. Inflammation 2021; 44:1895-1907. [PMID: 33860870 DOI: 10.1007/s10753-021-01466-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/19/2021] [Accepted: 04/05/2021] [Indexed: 11/25/2022]
Abstract
Asthma is an inflammatory airway disease wherein bronchoconstriction, airway inflammation, and airway obstruction during asthma attacks are the main problems. It is recognized that imbalance of Th1/Th2 and Th17/Treg is a critical factor in asthma pathogenesis. Manipulation of these with signaling molecules such as mTOR, PI3K, Akt, and MyD88 can control asthma. Mouse model of allergic asthma was produced and treated with ketamine, metformin, metformin and ketamine, triciribine, LY294002, and torin2. MCh challenge test, BALf's Eos Count, the IL-4, 5, INF-γ, eicosanoid, total IgE levels were determined. The MUC5a, Foxp3, RORγt, PI3K, mTOR, Akt, PU.1, and MyD88 gene expressions and histopathology study were done. Asthma groups that were treated with all six components had reduced Penh value, total IgE, IL-4 and IL-5 levels, MUC5a, RORγt, MyD88 and mTOR expression, goblet cell hyperplasia, and mucus hyper-secretion. The eosinophil percentage and Cys-LT level were decreased by metformin and ketamine, triciribine, LY294002, and torin2. The level of IFN-γ was increased in triciribine, LY294002, and torin2. Metformin, metformin and ketamine, triciribine, LY294002, and torin2 reduced Akt and PI3K expression, peribronchial and perivascular inflammation, and increased expression of Foxp3. Torin2 had an effect on PU.1 expression. Inhibition of PI3K/AKT/mTOR and TLR4/MyD88/NF-κB signaling with targeted molecules can attenuate asthma pathology and play an important role in airways protection.
Collapse
Affiliation(s)
- Baowei Ma
- Department of Thoracic Surgery, Xilingol League Hospital, Xilin Hot City, 026000, Inner Mongolia, China
| | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Entezar Mehrabi Nasab
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Limin Zhao
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
49
|
You YN, Xing QQ, Zhao X, Ji JJ, Yan H, Zhou T, Dong YM, Ren LS, Hou ST, Ding YY. Gu-Ben-Fang-Xiao decoction modulates lipid metabolism by activating the AMPK pathway in asthma remission. Biomed Pharmacother 2021; 138:111403. [PMID: 33714782 DOI: 10.1016/j.biopha.2021.111403] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/01/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Gu-Ben-Fang-Xiao decoction (GBFXD), derived from the traditional Chinese medicine Yu-Ping-Feng-San, is widely used in clinical settings and has obvious curative effects in respiratory diseases. GBFXD regulates cholesterol transport and lipid metabolism in chronic persistent asthma. There is evidence for its beneficial effects in the remission stage of asthma; however, its metabolic regulatory effects and underlying mechanisms during asthma remission are unclear. In the present study, we used liquid chromatography-mass spectrometry (LC-MS) to analyse the metabolic profile of mouse serum during asthma remission. The acquired LC-MS data were subjected to a multivariate analysis for identification of significantly altered metabolites. In total, 42 metabolites were significantly differentially expressed among the control, model, and GBFXD groups. In particular, levels of fatty acids, acylcarnitines, phosphatidylcholines, phosphatidylethanolamines, phosphatidylinositols, triglycerides, and diacylglycerols were altered during asthma remission. GBFXD may maintain lipid homeostasis on the lung surface by modulating lipid metabolism and may thereby alleviate asthma. We further quantified hypogeic acid (FA 16:1) based on targeted metabolomics and found that GBFXD may regulate fatty acid metabolism by activating the AMP-activated protein kinase (AMPK) pathway. These results support the use of GBFXD in patients with asthma remission.
Collapse
Affiliation(s)
- Yan-Nan You
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qiong-Qiong Xing
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xia Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jian-Jian Ji
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hua Yan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tao Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Ying-Mei Dong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li-Shun Ren
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shu-Ting Hou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuan-Yuan Ding
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China; Pediatric Institution of Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
50
|
Wu TD, Fawzy A, Kinney GL, Bon J, Neupane M, Tejwani V, Hansel NN, Wise RA, Putcha N, McCormack MC. Metformin use and respiratory outcomes in asthma-COPD overlap. Respir Res 2021; 22:70. [PMID: 33637087 PMCID: PMC7908718 DOI: 10.1186/s12931-021-01658-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/08/2021] [Indexed: 01/09/2023] Open
Abstract
Background Metformin is associated with improved respiratory outcomes in asthma; however, metformin in COPD and asthma-COPD overlap (ACO) remains unexplored. Objective To determine the association between metformin use and respiratory outcomes in COPD and ACO. Study design and methods Participants with COPD (FEV1/FVC < 0.70) in the Genetic Epidemiology of COPD study (COPDGene®) were categorized as ACO (n = 510), defined as concurrent physician-diagnosed asthma before age 40 years, or COPD alone (n = 3459). We estimated the association of baseline metformin use with (1) rate of total and severe respiratory exacerbations during follow-up, (2) cross-sectional St. George’s Respiratory Questionnaire (SGRQ) score, six-minute walk distance (6MWD), and post-bronchodilator FEV1 percent predicted (FEV1pp), and (3) 5-year change in SGRQ, 6MWD, and FEV1pp. We also examined change in SGRQ, 6MWD and FEV1pp among participants who initiated metformin during follow-up (n = 108) compared to persistent metformin non-users (n = 2080). Analyses were adjusted for sociodemographic factors, medications, and comorbidities. Results Among participants with ACO, metformin use was associated with lower rate of total (adjusted incidence rate ratio [aIRR] 0.3; 95% confidence interval [95%CI] 0.11, 0.77) and severe exacerbations (aIRR 0.29; 95%CI 0.10, 0.89). Among participants with COPD alone, there was no association between metformin use with total (aIRR 0.98; 95%CI 0.62, 1.5) or severe exacerbations (aIRR 1.3; 95% CI 0.68, 2.4) (p-interaction < 0.05). Metformin use was associated with lower baseline SGRQ score (adjusted mean difference [aMD] − 2.7; 95%CI − 5.3, − 0.2) overall. Metformin initiation was associated with improved SGRQ score (aMD –10.0; 95% CI − 18.7, − 1.2) among participants with ACO but not COPD alone (p-interaction < 0.05). There was no association between metformin use and 6MWD or FEV1pp in any comparison. Conclusions Metformin use was associated with fewer respiratory exacerbations and improved quality of life among individuals with ACO but not COPD alone. Results suggest a potential role for metformin in ACO which requires further prospective study. Trial Registry: NCT00608764
Collapse
Affiliation(s)
- Tianshi David Wu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA.,Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, Houston, TX, USA.,Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St. 5th Floor, Baltimore, MD, 21205, USA
| | - Ashraf Fawzy
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St. 5th Floor, Baltimore, MD, 21205, USA
| | - Gregory L Kinney
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Jessica Bon
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Maniraj Neupane
- Department of Critical Care Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Vickram Tejwani
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St. 5th Floor, Baltimore, MD, 21205, USA
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St. 5th Floor, Baltimore, MD, 21205, USA
| | - Robert A Wise
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St. 5th Floor, Baltimore, MD, 21205, USA
| | - Nirupama Putcha
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St. 5th Floor, Baltimore, MD, 21205, USA
| | - Meredith C McCormack
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, 1830 E. Monument St. 5th Floor, Baltimore, MD, 21205, USA.
| |
Collapse
|