1
|
Drake LY, Roos BB, Teske JJ, Borkar NA, Ayyalasomayajula S, Klapperich C, Koloko Ngassie ML, Pabelick CM, Prakash YS. Effects of glial-derived neurotrophic factor on remodeling and mitochondrial function in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2024; 327:L684-L693. [PMID: 39316680 DOI: 10.1152/ajplung.00101.2024] [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: 03/20/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/26/2024] Open
Abstract
Airway smooth muscle (ASM) cells play important roles in airway remodeling of asthma. Our previous studies show that in vivo administration of glial-derived neurotrophic factor (GDNF) in mice induces thickening and collagen deposition in bronchial airways, whereas chelation of GDNF by GFRα1-Fc attenuates airway remodeling in the context of allergen exposure. To determine whether GDNF has direct effects on ASM, in this study, we examined GDNF in ASM cells from normal versus asthmatic humans. We found that GDNF treatment of human ASM cells had only minor effects on cell proliferation and migration, intracellular expression or extracellular deposition of collagen I (COL1), collagen III (COL3), and fibronectin. Endoplasmic reticulum (ER) stress response and mitochondrial function have been implicated in asthma. We investigated whether GDNF regulates these aspects in human ASM. We found that GDNF treatment did not affect ER stress protein expression in normal or asthmatic cells. However, GDNF treatment impaired mitochondrial morphology in ASM but without significant effects on mitochondrial respiration. Thus, it is likely that in vivo effects of GDNF on airway remodeling per se involve cell types other than those on ASM, and thus ASM may serve more as a source of GDNF rather than a target.NEW & NOTEWORTHY Our previous study suggests that glial-derived neurotrophic factor (GDNF) is involved in allergen-induced airway hyperreactivity and remodeling in vivo. Here, we show that GDNF has no direct effects in remodeling of human airway smooth muscle (ASM) but GDNF dysregulates mitochondrial morphology in human ASM in the context of asthma.
Collapse
Affiliation(s)
- Li Y Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Benjamin B Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Jacob J Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Niyati A Borkar
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Savita Ayyalasomayajula
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Courtney Klapperich
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | | | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
2
|
Nopsopon T, Chen Y, Chen Q, Wheelock CE, Weiss ST, McGeachie M, Lasky-Su J, Akenroye A. Untargeted metabolomic analysis reveals different metabolites associated with response to mepolizumab and omalizumab in asthma. ERJ Open Res 2024; 10:00931-2023. [PMID: 39104961 PMCID: PMC11298997 DOI: 10.1183/23120541.00931-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/31/2024] [Indexed: 08/07/2024] Open
Abstract
Background There is limited evidence on biomarkers associated with response to the monoclonal antibodies currently approved for asthma treatment. We sought to identify circulatory metabolites associated with response to treatment with mepolizumab or omalizumab. Methods We conducted global metabolomic profiling of pre-treatment plasma samples from 100 patients with moderate-to-severe asthma who initiated mepolizumab (n=31) or omalizumab (n=69). The primary outcome was the change in exacerbations within 12 months of therapy. Negative binomial models were used to assess the association between each metabolite and exacerbations, adjusting for age, sex, body mass index, baseline exacerbations and inhaled corticosteroid use. Chemical similarity enrichment analysis (ChemRICH) was conducted to identify chemical subclasses associated with treatment response. Results The mean age of the mepolizumab group was 58.7 years with on average 2.9 exacerbations over the year prior to initiation of biologic therapy. The mean age in the omalizumab group was 48.8 years with 1.5 exacerbations in the preceding year. Patients with higher levels of two tocopherol metabolites were associated with more exacerbations on mepolizumab (δ-carboxyethyl hydroxychroman (CEHC) (p=2.65E-05, false discovery rate (FDR=0.01) and δ-CEHC glucuronide (p=2.47E-06, FDR=0.003)). Higher levels of six androgenic steroids, three carnitine metabolites and two bile acid metabolites were associated with decreased exacerbations in the omalizumab group. In enrichment analyses, xanthine metabolites (cluster FDR=0.0006) and tocopherol metabolites (cluster FDR=0.02) were associated with worse mepolizumab response, while androgenic steroids (cluster FDR=1.9E-18), pregnenolone steroids (cluster p=3.2E-07, FDR=1.4E-05) and secondary bile acid metabolites (cluster p=0.0003, FDR=0.006) were the top subclasses associated with better omalizumab response. Conclusion This study identifies distinct metabolites associated with response to mepolizumab and omalizumab, with androgenic steroids associated with response to both mepolizumab and omalizumab.
Collapse
Affiliation(s)
- Tanawin Nopsopon
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yulu Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Qingwen Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Craig E. Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Scott. T. Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael McGeachie
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- These authors contributed equally
| | - Ayobami Akenroye
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- These authors contributed equally
| |
Collapse
|
3
|
Chudakov DB, Shustova OA, Kotsareva OD, Generalov AA, Streltsova MS, Vavilova YD, Fattakhova GV. Chemical chaperone TUDCA selectively inhibits production of allergen-specific IgE in a low-dose model of allergy. BIOMEDITSINSKAIA KHIMIIA 2024; 70:5-14. [PMID: 38450676 DOI: 10.18097/pbmc20247001005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The cellular response to endoplasmic reticulum (ER) stress accompanies plasma cell maturation and is one of triggers and cofactors of the local inflammatory response. Chemical chaperones, low-molecular substances that eliminate pathological ER stress, are proposed as means of treating pathologies associated with ER stress. The aim of this study was to evaluate the effect and mechanisms of influence of chemical chaperones on the humoral response in a low-dose model of allergy. The allergic immune response was induced in BALB/c mice by repeated administration of ovalbumin at a dose of 100 ng for 6 weeks. Some animals were injected with both the antigen and the chemical chaperones, TUDCA (tauroursodeoxycholic acid) or 4-PBA (4-phenylbutyrate). Administration of TUDCA, but not 4-PBA, suppressed production of allergen-specific IgE (a 2.5-fold decrease in titer). None of the chemical chaperones affected the production of specific IgG1. The effect of TUDCA was associated with suppression of the switch to IgE synthesis in regional lymph nodes. This phenomenon was associated with suppressed expression of genes encoding cytokines involved in type 2 immune response, especially Il4 and Il9, which in turn could be caused by suppression of IL-33 release. In addition, TUDCA significantly suppressed expression of the cytokine APRIL, and to a lesser extent, BAFF. Thus, TUDCA inhibition of the allergy-specific IgE production is due to suppression of the release of IL-33 and a decrease in the production of type 2 immune response cytokines, as well as suppression of the expression of the cytokines APRIL and BAFF.
Collapse
Affiliation(s)
- D B Chudakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - O A Shustova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - O D Kotsareva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - A A Generalov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - M S Streltsova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Yu D Vavilova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - G V Fattakhova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
4
|
Hu L, Xu C, Tang X, Yu S, Wang L, Li Q, Zhou X. Fine particulate matter promotes airway inflammation and mucin production by activating endoplasmic reticulum stress and the IRE1α/NOD1/NF‑κB pathway. Int J Mol Med 2023; 52:96. [PMID: 37654182 PMCID: PMC10555484 DOI: 10.3892/ijmm.2023.5299] [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: 05/31/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
Fine particulate matter (PM2.5) is a type of small particle that is <2.5 µm in diameter that may cause airway inflammation. Thus, the present study aimed to explore the effects of PM2.5 on endoplasmic reticulum (ER) stress and airway inflammation in human airway epithelial cells. For this purpose, HBE135‑E6E7 airway epithelial cells were cultured and exposed to specific concentrations of PM2.5 for various periods of time, and cell viability was determined using a Cell Counting Kit‑8 assay. The results of the present study demonstrated that exposure to PM2.5 increased the mRNA and protein expression levels of interleukin (IL)‑6, tumor necrosis factor (TNF)‑α and mucin 5AC (MUC5AC). Moreover, the expression levels of ER stress‑related proteins, such as glucose‑regulated protein 78, CCAAT‑enhancer binding protein homologous protein, activating transcription factor 6, protein kinase R‑like ER kinase (PERK), phosphorylated (p‑)PERK, inositol‑requiring enzyme 1α (IRE1α) and p‑IRE1α, and nucleotide‑binding oligomerization domain 1 (NOD1) expression levels were increased following exposure to PM2.5. Transfection with IRE1α small interfering RNA (siRNA) led to the increased production of IL‑6, TNF‑α and MUC5AC. Moreover, the expression of NOD1 and the translocation of NF‑κB p65 were inhibited following transfection with IRE1α siRNA. In addition, the results of the present study demonstrated that transfection with NOD1 siRNA decreased the production of IL‑6, TNF‑α and MUC5AC, and decreased the translocation of NF‑κB p65. The expression levels of IL‑6, TNF‑α and MUC5AC were increased in the HBE135‑E6E7 cells following treatment with C12‑iE‑DAP, a NOD1 agonist. Moreover, treatment with C12‑iE‑DAP led to the activation of NF‑κB p65. Collectively, the results of the present study suggest that PM2.5 promotes airway inflammation and mucin production by activating ER stress in HBE135‑E6E7 airway epithelial cells, and that the IRE1α/NOD1/NF‑κB pathway may be involved in this process.
Collapse
Affiliation(s)
- Lihua Hu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Chaoqun Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Emergency and Trauma College, Hainan Medical University, Haikou, Hainan 579199, P.R. China
| | - Xiang Tang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Shanjun Yu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Lijun Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan 570102
| |
Collapse
|
5
|
Bruno S, Lamberty A, McCoy M, Mark Z, Daphtary N, Aliyeva M, Butnor K, Poynter ME, Anathy V, Cunniff B. Deletion of Miro1 in airway club cells potentiates allergic asthma phenotypes. FRONTIERS IN ALLERGY 2023; 4:1187945. [PMID: 37377691 PMCID: PMC10291198 DOI: 10.3389/falgy.2023.1187945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Mitochondria are multifaceted organelles necessary for numerous cellular signaling and regulatory processes. Mitochondria are dynamic organelles, trafficked and anchored to subcellular sites depending upon the cellular and tissue requirements. Precise localization of mitochondria to apical and basolateral membranes in lung epithelial cells is important for key mitochondrial processes. Miro1 is an outer mitochondrial membrane GTPase that associates with adapter proteins and microtubule motors to promote intracellular movement of mitochondria. We show that deletion of Miro1 in lung epithelial cells leads to perinuclear clustering of mitochondria. However, the role of Miro1 in epithelial cell response to allergic insults remains unknown. We generated a conditional mouse model to delete Miro1 in Club Cell Secretory Protein (CCSP) positive lung epithelial cells to examine the potential roles of Miro1 and mitochondrial trafficking in the lung epithelial response to the allergen, house dust mite (HDM). Our data show that Miro1 suppresses epithelial induction and maintenance of the inflammatory response to allergen, as Miro1 deletion modestly induces increases in pro-inflammatory signaling, specifically IL-6, IL-33, CCL20 and eotaxin levels, tissue reorganization, and airway hyperresponsiveness. Furthermore, loss of Miro1 in CCSP+ lung epithelial cells blocks resolution of the asthmatic insult. This study further demonstrates the important contribution of mitochondrial dynamic processes to the airway epithelial allergen response and the pathophysiology of allergic asthma.
Collapse
Affiliation(s)
- Sierra Bruno
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States
| | - Amelia Lamberty
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States
| | - Margaret McCoy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States
| | - Zoe Mark
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States
| | - Nirav Daphtary
- Department of Medicine, University of Vermont, Burlington, VT, United States
| | - Minara Aliyeva
- Department of Medicine, University of Vermont, Burlington, VT, United States
| | - Kelly Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States
| | - Matthew E. Poynter
- Department of Medicine, University of Vermont, Burlington, VT, United States
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
6
|
Bisgaard H, Chawes B, Stokholm J, Mikkelsen M, Schoos AMM, Bønnelykke K. 25 Years of translational research in the Copenhagen Prospective Studies on Asthma in Childhood (COPSAC). J Allergy Clin Immunol 2023; 151:619-633. [PMID: 36642652 DOI: 10.1016/j.jaci.2022.11.022] [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: 06/07/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 01/15/2023]
Abstract
The Copenhagen Prospective Studies on Asthma in Childhood (COPSAC) mother-child cohorts have provided a foundation of 25 years of research on the origins, prevention, and natural history of childhood asthma and related disorders. COPSAC's approach is characterized by clinical translational research with longitudinal deep phenotyping and exposure assessments from pregnancy, in combination with multi-omic data layers and embedded randomized controlled trials. One trial showed that fish oil supplementation during pregnancy prevented childhood asthma and identified pregnant women with the highest benefits from supplementation, thereby creating the potential for personalized prevention. COPSAC revealed that airway colonization with pathogenic bacteria in early life is associated with an increased risk of asthma. Further, airway bacteria were shown to be a trigger of acute asthma-like symptoms, with benefit from antibiotic treatment. COPSAC identified an immature gut microbiome in early life as a risk factor for asthma and allergy and further demonstrated that asthma can be predicted by infant lung function. At a molecular level, COPSAC has identified novel susceptibility genes, early immune deviations, and metabolomic alterations associated with childhood asthma. Thus, the COPSAC research program has enhanced our understanding of the processes causing childhood asthma and has suggested means of personalized prevention and treatment.
Collapse
Affiliation(s)
- Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Marianne Mikkelsen
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Marie Malby Schoos
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
7
|
Drake LY, Koloko Ngassie ML, Roos BB, Teske JJ, Prakash YS. Asthmatic lung fibroblasts promote type 2 immune responses via endoplasmic reticulum stress response dependent thymic stromal lymphopoietin secretion. Front Physiol 2023; 14:1064822. [PMID: 36760534 PMCID: PMC9907026 DOI: 10.3389/fphys.2023.1064822] [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: 10/08/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Lung fibroblasts contribute to asthma pathology partly through modulation of the immune environment in the airway. Tumor necrosis factor-α (TNFα) expression is upregulated in asthmatic lungs. How asthmatic lung fibroblasts respond to TNFα stimulation and subsequently regulate immune responses is not well understood. Endoplasmic reticulum (ER) stress and unfolded protein responses (UPR) play important roles in asthma, but their functional roles are still under investigation. In this study, we investigated TNFα-induced cytokine production in primary lung fibroblasts from asthmatic vs. non-asthmatic human subjects, and downstream effects on type 2 immune responses. TNFα significantly upregulated IL-6, IL-8, C-C motif chemokine ligand 5 (CCL5), and thymic stromal lymphopoietin (TSLP) mRNA expression and protein secretion by lung fibroblasts. Asthmatic lung fibroblasts secreted higher levels of TSLP which promoted IL-33-induced IL-5 and IL-13 production by peripheral blood mononuclear cells. TNFα exposure enhanced expression of ER stress/UPR pathways in both asthmatic and non-asthmatic lung fibroblasts, especially inositol-requiring protein 1α in asthmatics. ER stress/UPR inhibitors decreased IL-6, CCL5, and TSLP protein secretion by asthmatic lung fibroblasts. Our data suggest that TNFα and lung fibroblasts form an important axis in asthmatic lungs to promote asthmatic inflammation that can be attenuated by inhibiting ER stress/UPR pathway.
Collapse
Affiliation(s)
- Li Y. Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States,*Correspondence: Li Y. Drake,
| | - Maunick Lefin Koloko Ngassie
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands,Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Benjamin B. Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jacob J. Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Y. S. Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
8
|
Duan Q, Zhou Y, Yang D. Endoplasmic reticulum stress in airway hyperresponsiveness. Biomed Pharmacother 2022; 149:112904. [PMID: 35367759 DOI: 10.1016/j.biopha.2022.112904] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/18/2022] [Accepted: 03/27/2022] [Indexed: 11/15/2022] Open
Abstract
Airway hyperresponsiveness(AHR) is a major clinical phenomenon in lung diseases (asthma, COPD and pulmonary fibrosis) and not only a high-risk factor for perioperative airway spasm leading to hypoxaemia, haemodynamic instability and even "silent lung", but also a potential risk for increased mortality from underlying diseases (e.g. asthma, COPD). Airway reactivity is closely linked to airway inflammation, remodelling and increased mucus secretion, and endoplasmic reticulum stress is an important mechanism for the development of these pathologies. This review, therefore, focuses on the effects of endoplasmic reticulum stress on the immune cells involved in airway hyperreactivity (epithelial cells, dendritic cells, eosinophils and neutrophils) in inflammation and mucus & sputum secretion; and on the differentiation and remodelling of airway smooth muscle cells and epithelial cells. The aim is to clarify the mechanisms associated with endoplasmic reticulum stress in airway hyperresponsiveness and to find new ideas and methods for the prevention of airway hyperresponsiveness in the perioperative period.
Collapse
Affiliation(s)
- Qirui Duan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China
| | - Ying Zhou
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China
| | - Dong Yang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China.
| |
Collapse
|
9
|
Wang S, Gong L, Mo Y, Zhang J, Jiang Z, Tian Z, Shao C. Resveratrol attenuates inflammation and apoptosis through alleviating endoplasmic reticulum stress via Akt/mTOR pathway in fungus-induced allergic airways inflammation. Int Immunopharmacol 2021; 103:108489. [PMID: 34968999 DOI: 10.1016/j.intimp.2021.108489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/14/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Resveratrol has shown pleiotropic effects against inflammation and oxidative response. The present study aimed to investigate the effects and mechanisms of resveratrol on fungus-induced allergic airway inflammation. METHODS Female BALB/c mice were injected intraperitoneally with Aspergillus fumigatus (Af) extract emulsified with aluminum on day 0 and 7 and intranasally challenged with Af extracts on day 14 and 15. Resveratrol or dexamethasone or a vehicle was injected intraperitoneally 1 h before each challenge. Mice were sacrificed for serum, bronchoalveolar lavage fluid (BALF), and lungs 24 h after the last challenge. The control group was administered with saline. BEAS-2B was used for the experiments in vitro that Af-exposed airway epithelial cells. RESULTS Resveratrol and dexamethasone attenuated the airway inflammation and eosinophilia, and reduced not only the production of IL-4, IL-5, and IL-13 in the BALF and lung tissues but also the mRNA levels of lung IL-6, TNF-α, and TGF-β induced by Af challenge (P < 0.05). Furthermore, Af-induced lung endoplasmic reticulum (ER) stress-related proteins PERK, CHOP, and GRP78 and the apoptosis markers including cleaved caspase-3 and cleaved caspase-7 were both suppressed significantly by resveratrol (P < 0.05). In vitro, activation of ER stress and the Akt/mTOR pathway in Af-exposed BEAS-2B cells were effectively ameliorated by resveratrol. Inhibition of the Akt/mTOR pathway using LY294002 suppressed the ER stress while ER stress inhibitor 4-PBA decreased the apoptosis in Af-exposed BEAS-2B cells. CONCLUSIONS Our findings collectively revealed that resveratrol alleviated the Af-exposed allergic inflammation and apoptosis through inhibiting ER stress via Akt/mTOR pathway, exerting therapeutic effects on the fungus-induced allergic lung disorder.
Collapse
Affiliation(s)
- Sijiao Wang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Linjing Gong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuqing Mo
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Zhang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhilong Jiang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhengan Tian
- Shanghai International Travel Health Care Center, Shanghai 200335, China
| | - Changzhou Shao
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Pulmonary Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen 361015, China.
| |
Collapse
|
10
|
Nakada EM, Sun R, Fujii U, Martin JG. The Impact of Endoplasmic Reticulum-Associated Protein Modifications, Folding and Degradation on Lung Structure and Function. Front Physiol 2021; 12:665622. [PMID: 34122136 PMCID: PMC8188853 DOI: 10.3389/fphys.2021.665622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
The accumulation of unfolded/misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and induces the unfolded protein response (UPR) and other mechanisms to restore ER homeostasis, including translational shutdown, increased targeting of mRNAs for degradation by the IRE1-dependent decay pathway, selective translation of proteins that contribute to the protein folding capacity of the ER, and activation of the ER-associated degradation machinery. When ER stress is excessive or prolonged and these mechanisms fail to restore proteostasis, the UPR triggers the cell to undergo apoptosis. This review also examines the overlooked role of post-translational modifications and their roles in protein processing and effects on ER stress and the UPR. Finally, these effects are examined in the context of lung structure, function, and disease.
Collapse
Affiliation(s)
- Emily M. Nakada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Rui Sun
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Utako Fujii
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - James G. Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| |
Collapse
|
11
|
Khomari F, Nabi-Afjadi M, Yarahmadi S, Eskandari H, Bahreini E. Effects of Cell Proteostasis Network on the Survival of SARS-CoV-2. Biol Proced Online 2021; 23:8. [PMID: 33618659 PMCID: PMC7899210 DOI: 10.1186/s12575-021-00145-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/09/2021] [Indexed: 01/18/2023] Open
Abstract
The proteostasis network includes all the factors that control the function of proteins in their native state and minimize their non-functional or harmful reactions. The molecular chaperones, the important mediator in the proteostasis network can be considered as any protein that contributes to proper folding and assembly of other macromolecules, through maturating of unfolded or partially folded macromolecules, refolding of stress-denatured proteins, and modifying oligomeric assembly, otherwise it leads to their proteolytic degradation. Viruses that use the hosts' gene expression tools and protein synthesis apparatus to survive and replicate, are obviously protected by such a host chaperone system. This means that many viruses use members of the hosts' chaperoning system to infect the target cells, replicate, and spread. During viral infection, increase in endoplasmic reticulum (ER) stress due to high expression of viral proteins enhances the level of heat shock proteins (HSPs) and induces cell apoptosis or necrosis. Indeed, evidence suggests that ER stress and the induction of unfolded protein response (UPR) may be a major aspect of the corona-host virus interaction. In addition, several clinical reports have confirmed the autoimmune phenomena in COVID-19-patients, and a strong association between this autoimmunity and severe SARS-CoV-2 infection. Part of such autoimmunity is due to shared epitopes among the virus and host. This article reviews the proteostasis network and its relationship to the immune system in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Fateme Khomari
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Sahar Yarahmadi
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran
| | - Hanie Eskandari
- Department of Biology, Science and Research Branch, Islamic Azad University of Tehran, Tehran, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran
| |
Collapse
|
12
|
Chong WC, Shastri MD, Peterson GM, Patel RP, Pathinayake PS, Dua K, Hansbro NG, Hsu AC, Wark PA, Shukla SD, Johansen MD, Schroder K, Hansbro PM. The complex interplay between endoplasmic reticulum stress and the NLRP3 inflammasome: a potential therapeutic target for inflammatory disorders. Clin Transl Immunology 2021; 10:e1247. [PMID: 33614031 PMCID: PMC7878118 DOI: 10.1002/cti2.1247] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/09/2021] [Accepted: 01/10/2021] [Indexed: 12/15/2022] Open
Abstract
Inflammation is the result of a complex network of cellular and molecular interactions and mechanisms that facilitate immune protection against intrinsic and extrinsic stimuli, particularly pathogens, to maintain homeostasis and promote tissue healing. However, dysregulation in the immune system elicits excess/abnormal inflammation resulting in unintended tissue damage and causes major inflammatory diseases including asthma, chronic obstructive pulmonary disease, atherosclerosis, inflammatory bowel diseases, sarcoidosis and rheumatoid arthritis. It is now widely accepted that both endoplasmic reticulum (ER) stress and inflammasomes play critical roles in activating inflammatory signalling cascades. Notably, evidence is mounting for the involvement of ER stress in exacerbating inflammasome-induced inflammatory cascades, which may provide a new axis for therapeutic targeting in a range of inflammatory disorders. Here, we comprehensively review the roles, mechanisms and interactions of both ER stress and inflammasomes, as well as their interconnected relationships in inflammatory signalling cascades. We also discuss novel therapeutic strategies that are being developed to treat ER stress- and inflammasome-related inflammatory disorders.
Collapse
Affiliation(s)
- Wai Chin Chong
- Department of Molecular and Translational ScienceMonash UniversityClaytonVICAustralia
- Centre for Cancer ResearchHudson Institute of Medical ResearchClaytonVICAustralia
| | - Madhur D Shastri
- School of Pharmacy and PharmacologyUniversity of TasmaniaHobartTASAustralia
| | - Gregory M Peterson
- School of Pharmacy and PharmacologyUniversity of TasmaniaHobartTASAustralia
| | - Rahul P Patel
- School of Pharmacy and PharmacologyUniversity of TasmaniaHobartTASAustralia
| | - Prabuddha S Pathinayake
- Priority Research Centre for Healthy LungsHunter Medical Research InstituteThe University of NewcastleCallaghanNSWAustralia
| | - Kamal Dua
- Discipline of PharmacyGraduate School of HealthUniversity of Technology SydneyUltimoNSWAustralia
| | - Nicole G Hansbro
- Centre for InflammationCentenary InstituteFaculty of ScienceSchool of Life SciencesUniversity of TechnologySydneyNSWAustralia
| | - Alan C Hsu
- Priority Research Centre for Healthy LungsHunter Medical Research InstituteThe University of NewcastleCallaghanNSWAustralia
| | - Peter A Wark
- Priority Research Centre for Healthy LungsHunter Medical Research InstituteThe University of NewcastleCallaghanNSWAustralia
| | - Shakti Dhar Shukla
- Priority Research Centre for Healthy LungsHunter Medical Research InstituteThe University of NewcastleCallaghanNSWAustralia
| | - Matt D Johansen
- Centre for InflammationCentenary InstituteFaculty of ScienceSchool of Life SciencesUniversity of TechnologySydneyNSWAustralia
| | - Kate Schroder
- Institute for Molecular BioscienceUniversity of QueenslandSt LuciaQLDAustralia
| | - Philip M Hansbro
- Priority Research Centre for Healthy LungsHunter Medical Research InstituteThe University of NewcastleCallaghanNSWAustralia
- Centre for InflammationCentenary InstituteFaculty of ScienceSchool of Life SciencesUniversity of TechnologySydneyNSWAustralia
| |
Collapse
|
13
|
Bradley KL, Stokes CA, Marciniak SJ, Parker LC, Condliffe AM. Role of unfolded proteins in lung disease. Thorax 2021; 76:92-99. [PMID: 33077618 PMCID: PMC7803888 DOI: 10.1136/thoraxjnl-2019-213738] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 01/01/2023]
Abstract
The lungs are exposed to a range of environmental toxins (including cigarette smoke, air pollution, asbestos) and pathogens (bacterial, viral and fungal), and most respiratory diseases are associated with local or systemic hypoxia. All of these adverse factors can trigger endoplasmic reticulum (ER) stress. The ER is a key intracellular site for synthesis of secretory and membrane proteins, regulating their folding, assembly into complexes, transport and degradation. Accumulation of misfolded proteins within the lumen results in ER stress, which activates the unfolded protein response (UPR). Effectors of the UPR temporarily reduce protein synthesis, while enhancing degradation of misfolded proteins and increasing the folding capacity of the ER. If successful, homeostasis is restored and protein synthesis resumes, but if ER stress persists, cell death pathways are activated. ER stress and the resulting UPR occur in a range of pulmonary insults and the outcome plays an important role in many respiratory diseases. The UPR is triggered in the airway of patients with several respiratory diseases and in corresponding experimental models. ER stress has been implicated in the initiation and progression of pulmonary fibrosis, and evidence is accumulating suggesting that ER stress occurs in obstructive lung diseases (particularly in asthma), in pulmonary infections (some viral infections and in the setting of the cystic fibrosis airway) and in lung cancer. While a number of small molecule inhibitors have been used to interrogate the role of the UPR in disease models, many of these tools have complex and off-target effects, hence additional evidence (eg, from genetic manipulation) may be required to support conclusions based on the impact of such pharmacological agents. Aberrant activation of the UPR may be linked to disease pathogenesis and progression, but at present, our understanding of the context-specific and disease-specific mechanisms linking these processes is incomplete. Despite this, the ability of the UPR to defend against ER stress and influence a range of respiratory diseases is becoming increasingly evident, and the UPR is therefore attracting attention as a prospective target for therapeutic intervention strategies.
Collapse
Affiliation(s)
- Kirsty L Bradley
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | - Clare A Stokes
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | | | - Lisa C Parker
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| |
Collapse
|
14
|
Dastghaib S, Kumar PS, Aftabi S, Damera G, Dalvand A, Sepanjnia A, Kiumarsi M, Aghanoori MR, Sohal SS, Ande SR, Alizadeh J, Mokarram P, Ghavami S, Sharma P, Zeki AA. Mechanisms Targeting the Unfolded Protein Response in Asthma. Am J Respir Cell Mol Biol 2021; 64:29-38. [PMID: 32915643 DOI: 10.1165/rcmb.2019-0235tr] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
Lung cells are constantly exposed to various internal and external stressors that disrupt protein homeostasis. To cope with these stimuli, cells evoke a highly conserved adaptive mechanism called the unfolded protein response (UPR). UPR stressors can impose greater protein secretory demands on the endoplasmic reticulum (ER), resulting in the development, differentiation, and survival of these cell types to meet these increasing functional needs. Dysregulation of the UPR leads to the development of the disease. The UPR and ER stress are involved in several human conditions, such as chronic inflammation, neurodegeneration, metabolic syndrome, and cancer. Furthermore, potent and specific compounds that target the UPR pathway are under development as future therapies. The focus of this review is to thoroughly describe the effects of both internal and external stressors on the ER in asthma. Furthermore, we discuss how the UPR signaling pathway is activated in the lungs to overcome cellular damage. We also present an overview of the pathogenic mechanisms, with a brief focus on potential strategies for pharmacological interventions.
Collapse
Affiliation(s)
- Sanaz Dastghaib
- Department of Clinical Biochemistry and
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - P Sravan Kumar
- National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Sajjad Aftabi
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
- Medical Physics Department and
| | - Gautam Damera
- Personalized and Predictive Medicine (Respiratory), Global Research and Development, Teva Pharmaceuticals, Malvern, Pennsylvania
| | - Azadeh Dalvand
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
| | - Adel Sepanjnia
- Department of Immunology, School of Medicine, Jiroft University of Medical Science, Jiroft, Iran
| | - Mohammad Kiumarsi
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
| | - Mohamad-Reza Aghanoori
- Department of Human Genetics, School of Medicine, and
- Department of Pharmacology and Therapeutics
- Division of Neurodegenerative Disorders, Albrechtsen Research Centre, St. Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | | | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
- Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pooneh Mokarram
- Department of Clinical Biochemistry and
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Ghavami
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine
- Department of Internal Medicine, and
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan Sharma
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Amir A Zeki
- Lung Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, California; and
- Veterans Affairs Medical Center, Mather, California
| |
Collapse
|
15
|
Inflammation-Induced Protein Unfolding in Airway Smooth Muscle Triggers a Homeostatic Response in Mitochondria. Int J Mol Sci 2020; 22:ijms22010363. [PMID: 33396378 PMCID: PMC7795579 DOI: 10.3390/ijms22010363] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/17/2020] [Accepted: 12/28/2020] [Indexed: 12/11/2022] Open
Abstract
The effects of airway inflammation on airway smooth muscle (ASM) are mediated by pro-inflammatory cytokines such as tumor necrosis factor alpha (TNFα). In this review article, we will provide a unifying hypothesis for a homeostatic response to airway inflammation that mitigates oxidative stress and thereby provides resilience to ASM. Previous studies have shown that acute exposure to TNFα increases ASM force generation in response to muscarinic stimulation (hyper-reactivity) resulting in increased ATP consumption and increased tension cost. To meet this increased energetic demand, mitochondrial O2 consumption and oxidative phosphorylation increases but at the cost of increased reactive oxygen species (ROS) production (oxidative stress). TNFα-induced oxidative stress results in the accumulation of unfolded proteins in the endoplasmic reticulum (ER) and mitochondria of ASM. In the ER, TNFα selectively phosphorylates inositol-requiring enzyme 1 alpha (pIRE1α) triggering downstream splicing of the transcription factor X-box binding protein 1 (XBP1s); thus, activating the pIRE1α/XBP1s ER stress pathway. Protein unfolding in mitochondria also triggers an unfolded protein response (mtUPR). In our conceptual framework, we hypothesize that activation of these pathways is homeostatically directed towards mitochondrial remodeling via an increase in peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC1α) expression, which in turn triggers: (1) mitochondrial fragmentation (increased dynamin-related protein-1 (Drp1) and reduced mitofusin-2 (Mfn2) expression) and mitophagy (activation of the Phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1)/Parkin mitophagy pathway) to improve mitochondrial quality; (2) reduced Mfn2 also results in a disruption of mitochondrial tethering to the ER and reduced mitochondrial Ca2+ influx; and (3) mitochondrial biogenesis and increased mitochondrial volume density. The homeostatic remodeling of mitochondria results in more efficient O2 consumption and oxidative phosphorylation and reduced ROS formation by individual mitochondrion, while still meeting the increased ATP demand. Thus, the energetic load of hyper-reactivity is shared across the mitochondrial pool within ASM cells.
Collapse
|
16
|
Miao K, Zhang L, Pan T, Wang Y. Update on the role of endoplasmic reticulum stress in asthma. Am J Transl Res 2020; 12:1168-1183. [PMID: 32355534 PMCID: PMC7191165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/23/2020] [Indexed: 06/11/2023]
Abstract
Asthma has long attracted extensive attention because of its recurring symptoms of reversible airflow obstruction, airway hyperresponsiveness (AHR) and airway inflammation. Although accumulating evidence has enabled gradual increases in understanding of the pathogenesis of asthma, many questions regarding the mechanisms underlying asthma onset and progression remain unanswered. Recent advances delineating the potential functions of endoplasmic reticulum (ER) stress in meeting the need for an airway hypersensitivity response have revealed critical roles of unfolded protein response (UPR) pathways in asthma. In this review, we highlight the roles of ER stress and UPR activation in the etiology, pathogenesis and treatment of asthma and discuss whether the related mechanisms could be targets for therapeutic strategies.
Collapse
Affiliation(s)
- Kang Miao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology 1095 Jiefang Ave, Wuhan 430030, China
| | - Lei Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology 1095 Jiefang Ave, Wuhan 430030, China
| | - Ting Pan
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology 1095 Jiefang Ave, Wuhan 430030, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology 1095 Jiefang Ave, Wuhan 430030, China
| |
Collapse
|
17
|
Delmotte P, Sieck GC. Endoplasmic Reticulum Stress and Mitochondrial Function in Airway Smooth Muscle. Front Cell Dev Biol 2020; 7:374. [PMID: 32010691 PMCID: PMC6974519 DOI: 10.3389/fcell.2019.00374] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/16/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammatory airway diseases such as asthma affect more than 300 million people world-wide. Inflammation triggers pathophysiology via such as tumor necrosis factor α (TNFα) and interleukins (e.g., IL-13). Hypercontraction of airway smooth muscle (ASM) and ASM cell proliferation are major contributors to the exaggerated airway narrowing that occurs during agonist stimulation. An emergent theme in this context is the role of inflammation-induced endoplasmic reticulum (ER) stress and altered mitochondrial function including an increase in the formation of reactive oxygen species (ROS). This may establish a vicious cycle as excess ROS generation leads to further ER stress. Yet, it is unclear whether inflammation-induced ROS is the major mechanism leading to ER stress or the consequence of ER stress. In various diseases, inflammation leads to an increase in mitochondrial fission (fragmentation), associated with reduced levels of mitochondrial fusion proteins, such as mitofusin 2 (Mfn2). Mitochondrial fragmentation may be a homeostatic response since it is generally coupled with mitochondrial biogenesis and mitochondrial volume density thereby reducing demand on individual mitochondrion. ER stress is triggered by the accumulation of unfolded proteins, which induces a homeostatic response to alter protein balance via effects on protein synthesis and degradation. In addition, the ER stress response promotes protein folding via increased expression of molecular chaperone proteins. Reduced Mfn2 and altered mitochondrial dynamics may not only be downstream to ER stress but also upstream such that a reduction in Mfn2 triggers further ER stress. In this review, we summarize the current understanding of the link between inflammation-induced ER stress and mitochondrial function and the role played in the pathophysiology of inflammatory airway diseases.
Collapse
Affiliation(s)
- Philippe Delmotte
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
18
|
Yoon YH, Yeon SH, Choi MR, Jang YS, Kim JA, Oh HW, Jun X, Park SK, Heo JY, Rha KS, Kim YM. Altered Mitochondrial Functions and Morphologies in Epithelial Cells Are Associated With Pathogenesis of Chronic Rhinosinusitis With Nasal Polyps. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:653-668. [PMID: 32400131 PMCID: PMC7224996 DOI: 10.4168/aair.2020.12.4.653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/16/2020] [Accepted: 02/25/2020] [Indexed: 12/29/2022]
Abstract
Purpose Chronic rhinosinusitis with nasal polyps (CRSwNP) is a complex inflammatory disease of the nasal and paranasal sinus mucosa. The disease is associated with mitochondrial dysfunction, structural changes in the mitochondria, and reactive oxygen species (ROS) generation. This study investigated whether there are functional and morphological changes in the mitochondria in the epithelial cells of nasal polyps (NPs) and Staphylococcus aureus enterotoxin B (SEB)-stimulated nasal epithelial cells. Methods In all, 30 patients with CRSwNP and 15 healthy subjects were enrolled. Mitochondrial ROS (mtROS) and changes in mitochondrial functions and structures were investigated in the uncinate tissue (UT) of healthy controls, the UT or NPs of CRSwNP patients, and human nasal epithelial cells with or without SEB stimulation. Results Oxidative phosphorylation complexes showed various responses following SEB stimulation in the nasal epithelial cells, and their expressions were significantly higher in the NPs of patients with CRSwNP than in the UT of controls. Generation of mtROS was increased following SEB exposure in nasal epithelial cells and was reduced by pretreatment with MitoTEMPO, which is used as an mtROS scavenger. In the tissues, mtROS was significantly increased in the NPs of CRSwNP patients compared to the UT of controls or CRSwNP patients. The expressions of fusion- and fission-related molecules were also significantly higher in SEB-exposed nasal epithelial cells than in non-exposed cells. In tissues, the expression of fission (fission mediator protein 1)- and fusion (membrane and mitofusin-1, and optic atrophy protein 1)-related molecules was significantly higher in the NPs of CRSwNP patients than in UT of controls or CRSwNP patients. Transmission electron microscopy revealed elongated mitochondria in SEB-exposed nasal epithelial cells and epithelial cells of NPs. Conclusions Production of mtROS, disrupted mitochondrial function, and structural changes in nasal epithelial cells might be involved in the pathogenesis of CRSwNP.
Collapse
Affiliation(s)
- Young Hoon Yoon
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sun Hee Yeon
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon, Korea
| | - Mi Ra Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Yoon Sun Jang
- Department of Biochemistry, Research Institute for Neurosciences, Chungnam National University School of Medicine, Daejeon, Korea
| | - Ji Ae Kim
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Hyun Woo Oh
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Xu Jun
- State Key Laboratory of Respiratory Disease, Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital, Guangzhou Medical University, Guanzhou, China
| | - Soo Kyung Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jun Young Heo
- Department of Biochemistry, Research Institute for Neurosciences, Chungnam National University School of Medicine, Daejeon, Korea
| | - Ki Sang Rha
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon, Korea
| | - Yong Min Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon, Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea.
| |
Collapse
|
19
|
Protein Misfolding and Endoplasmic Reticulum Stress in Chronic Lung Disease: Will Cell-Specific Targeting Be the Key to the Cure? Chest 2019; 157:1207-1220. [PMID: 31778676 DOI: 10.1016/j.chest.2019.11.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/07/2019] [Accepted: 11/10/2019] [Indexed: 12/31/2022] Open
Abstract
Chronic lung disease accounts for a significant global burden with respect to death, disability, and health-care costs. Due to the heterogeneous nature and limited treatment options for these diseases, it is imperative that the cellular and molecular mechanisms underlying the disease pathophysiology are further understood. The lung is a complex organ with a diverse cell population, and each cell type will likely have different roles in disease initiation, progression, and resolution. The effectiveness of a given therapeutic agent may depend on the net effect on each of these cell types. Over the past decade, it has been established that endoplasmic reticulum stress and the unfolded protein response are involved in the development of several chronic lung diseases. These conserved cellular pathways are important for maintaining cellular proteostasis, but their aberrant activation can result in pathology. This review discusses the current understanding of endoplasmic reticulum stress and the unfolded protein response at the cellular level in the development and progression of various chronic lung diseases. We highlight the need for increased understanding of the specific cellular contributions of unfolded protein response activation to these pathologies and suggest that the development of cell-specific targeted therapies is likely required to further decrease disease progression and to promote resolution of chronic lung disease.
Collapse
|
20
|
Kusaczuk M. Tauroursodeoxycholate-Bile Acid with Chaperoning Activity: Molecular and Cellular Effects and Therapeutic Perspectives. Cells 2019; 8:E1471. [PMID: 31757001 PMCID: PMC6952947 DOI: 10.3390/cells8121471] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/15/2019] [Accepted: 11/19/2019] [Indexed: 12/11/2022] Open
Abstract
Tauroursodeoxycholic acid (TUDCA) is a naturally occurring hydrophilic bile acid that has been used for centuries in Chinese medicine. Chemically, TUDCA is a taurine conjugate of ursodeoxycholic acid (UDCA), which in contemporary pharmacology is approved by Food and Drug Administration (FDA) for treatment of primary biliary cholangitis. Interestingly, numerous recent studies demonstrate that mechanisms of TUDCA functioning extend beyond hepatobiliary disorders. Thus, TUDCA has been demonstrated to display potential therapeutic benefits in various models of many diseases such as diabetes, obesity, and neurodegenerative diseases, mostly due to its cytoprotective effect. The mechanisms underlying this cytoprotective activity have been mainly attributed to alleviation of endoplasmic reticulum (ER) stress and stabilization of the unfolded protein response (UPR), which contributed to naming TUDCA as a chemical chaperone. Apart from that, TUDCA has also been found to reduce oxidative stress, suppress apoptosis, and decrease inflammation in many in-vitro and in-vivo models of various diseases. The latest research suggests that TUDCA can also play a role as an epigenetic modulator and act as therapeutic agent in certain types of cancer. Nevertheless, despite the massive amount of evidence demonstrating positive effects of TUDCA in pre-clinical studies, there are certain limitations restraining its wide use in patients. Here, molecular and cellular modes of action of TUDCA are described and therapeutic opportunities and limitations of this bile acid are discussed.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Białystok, Mickiewicza 2A, 15-222 Białystok, Poland
| |
Collapse
|
21
|
Chawes BL, Giordano G, Pirillo P, Rago D, Rasmussen MA, Stokholm J, Bønnelykke K, Bisgaard H, Baraldi E. Neonatal Urine Metabolic Profiling and Development of Childhood Asthma. Metabolites 2019; 9:metabo9090185. [PMID: 31527391 PMCID: PMC6780518 DOI: 10.3390/metabo9090185] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022] Open
Abstract
Urine metabolomics case-control studies of childhood asthma have demonstrated a discriminative ability. Here, we investigated whether urine metabolic profiles from healthy neonates were associated with the development of asthma in childhood. Untargeted metabolomics by liquid chromatography-mass spectrometry was applied to urine samples collected at age 4 weeks in 171 and 161 healthy neonates born from mothers with asthma from the COPSAC2000 and COPSAC2010 cohorts, respectively, where persistent wheeze/asthma was prospectively diagnosed using a symptom-based algorithm. Univariate and multivariate analyses were applied to investigate differences in metabolic profiles between children who developed asthma and healthy children. Univariate analysis showed 63 and 87 metabolites (q-value < 0.15) in COPSAC2000 and COPSAC2010, respectively, which is promising for discriminating between asthmatic and healthy children. Of those, 14 metabolites were common among the two cohorts. Multivariate random forest and projection to latent structures discriminant analyses confirmed the discriminatory capacity of the metabolic profiles in both cohorts with estimated errors in prediction equal to 35% and AUCpred > 0.60. Database search enabled annotation of three discriminative features: a glucoronidated compound (steroid), 3-hydroxytetradecanedioic acid (fatty acid), and taurochenodeoxycholate-3-sulfate (bile acid). The urine metabolomics profiles from healthy neonates were associated with the development of childhood asthma, but further research is needed to understand underlying metabolic pathways.
Collapse
Affiliation(s)
- Bo L Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, 2820 Copenhagen, Denmark.
| | - Giuseppe Giordano
- Women's and Children's Health Department, University of Padova, 35128 Padova, Italy.
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35127 Padova, Italy.
| | - Paola Pirillo
- Women's and Children's Health Department, University of Padova, 35128 Padova, Italy.
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35127 Padova, Italy.
| | - Daniela Rago
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, 2820 Copenhagen, Denmark.
| | - Morten A Rasmussen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, 2820 Copenhagen, Denmark.
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, 2820 Copenhagen, Denmark.
- Department of Pediatrics, Naestved Hospital, 4700 Naestved, Denmark.
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, 2820 Copenhagen, Denmark.
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, 2820 Copenhagen, Denmark.
| | - Eugenio Baraldi
- Women's and Children's Health Department, University of Padova, 35128 Padova, Italy.
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35127 Padova, Italy.
| |
Collapse
|
22
|
Castro I, Albornoz N, Aguilera S, Barrera MJ, González S, Núñez M, Carvajal P, Jara D, Lagos C, Molina C, Urzúa U, Hermoso MA, González MJ. Aberrant MUC1 accumulation in salivary glands of Sjögren’s syndrome patients is reversed by TUDCA in vitro. Rheumatology (Oxford) 2019; 59:742-753. [DOI: 10.1093/rheumatology/kez316] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/28/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
Objectives
Xerostomia in SS patients has been associated with low quality and quantity of salivary mucins, which are fundamental for the hydration and protection of the oral mucosa. The aim of this study was to evaluate if cytokines induce aberrant mucin expression and whether tauroursodeoxycholic acid (TUDCA) is able to counteract such an anomaly.
Methods
Labial salivary glands from 16 SS patients and 15 control subjects, as well as 3D acini or human submandibular gland cells stimulated with TNF-α or IFN-γ and co-incubated with TUDCA, were analysed. mRNA and protein levels of Mucin 1 (MUC1) and MUC7 were determined by RT-qPCR and western blot, respectively. Co-immunoprecipitation and immunofluorescence assays for mucins and GRP78 [an endoplasmic reticulum (ER)-resident protein] were also performed. mRNA levels of RelA/p65 (nuclear factor-κB subunit), TNF-α, IL-1β, IL-6, SEL1L and EDEM1 were determined by RT-qPCR, and RelA/p65 localization was evaluated by immunofluorescence.
Results
MUC1 is overexpressed and accumulated in the ER of labial salivary gland from SS patients, while MUC7 accumulates throughout the cytoplasm of acinar cells; however, MUC1, but not MUC7, co-precipitated with GRP78. TUDCA diminished the overexpression and aberrant accumulation of MUC1 induced by TNF-α and IFN-γ, as well as the nuclear translocation of RelA/p65, together with the expression of inflammatory and ER stress markers in 3D acini.
Conclusion
Chronic inflammation alters the secretory process of MUC1, inducing ER stress and affecting the quality of saliva in SS patients. TUDCA showed anti-inflammatory properties decreasing aberrant MUC1 accumulation. Further studies are necessary to evaluate the potential therapeutic effect of TUDCA in restoring glandular homeostasis in SS patients.
Collapse
Affiliation(s)
| | - Nicolás Albornoz
- Programa de Biología Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sergio Aguilera
- Departamento de Reumatología, Clínica INDISA, Santiago, Chile
| | | | - Sergio González
- Escuela de Odontología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Matilde Núñez
- Programa de Biología Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Patricia Carvajal
- Programa de Biología Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Daniela Jara
- Programa de Biología Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Carolina Lagos
- Programa de Biología Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Claudio Molina
- Facultad de Odontología, Universidad San Sebastián, Santiago, Chile
| | - Ulises Urzúa
- Departamento de Oncología Básico-Clínico y, Santiago, Chile
| | - Marcela A Hermoso
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María-Julieta González
- Programa de Biología Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
23
|
Abstract
Asthma is a genetically and phenotypically complex disease that has a major impact on global health. Signs and symptoms of asthma are caused by the obstruction of airflow through the airways. The epithelium that lines the airways plays a major role in maintaining airway patency and in host defense. The epithelium initiates responses to inhaled or aspirated substances, including allergens, viruses, and bacteria, and epithelial-derived cytokines are important in the recruitment and activation of immune cells in the airway. Changes in the structure and function of the airway epithelium are a prominent feature of asthma. Approximately half of individuals with asthma have evidence of active type 2 immune responses in the airway. In these individuals, epithelial cytokines promote type 2 responses, and responses to type 2 cytokines result in increased epithelial mucus production and other effects that cause airway obstruction. Recent work also implicates other epithelial responses, including interleukin-17, interferon and ER stress responses, that may contribute to asthma pathogenesis and provide new targets for therapy.
Collapse
Affiliation(s)
- Luke R Bonser
- Lung Biology Center, University of California San Francisco, San Francisco, CA, United States
| | - David J Erle
- Lung Biology Center, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
24
|
Nakada EM, Bhakta NR, Korwin-Mihavics BR, Kumar A, Chamberlain N, Bruno SR, Chapman DG, Hoffman SM, Daphtary N, Aliyeva M, Irvin CG, Dixon AE, Woodruff PG, Amin S, Poynter ME, Desai DH, Anathy V. Conjugated bile acids attenuate allergen-induced airway inflammation and hyperresponsiveness by inhibiting UPR transducers. JCI Insight 2019; 4:98101. [PMID: 31045581 DOI: 10.1172/jci.insight.98101] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 04/02/2019] [Indexed: 12/14/2022] Open
Abstract
Conjugated bile acids (CBAs), such as tauroursodeoxycholic acid (TUDCA), are known to resolve the inflammatory and unfolded protein response (UPR) in inflammatory diseases, such as asthma. Whether CBAs exert their beneficial effects on allergic airway responses via 1 arm or several arms of the UPR, or alternatively through the signaling pathways for conserved bile acid receptor, remains largely unknown. We used a house dust mite-induced (HDM-induced) murine model of asthma to evaluate and compare the effects of 5 CBAs and 1 unconjugated bile acid in attenuating allergen-induced UPR and airway responses. Expression of UPR-associated transcripts was assessed in airway brushings from human patients with asthma and healthy subjects. Here we show that CBAs, such as alanyl β-muricholic acid (AβM) and TUDCA, significantly decreased inflammatory, immune, and cytokine responses; mucus metaplasia; and airway hyperresponsiveness, as compared with other CBAs in a model of allergic airway disease. CBAs predominantly bind to activating transcription factor 6α (ATF6α) compared with the other canonical transducers of the UPR, subsequently decreasing allergen-induced UPR activation and resolving allergic airway disease, without significant activation of the bile acid receptors. TUDCA and AβM also attenuated other HDM-induced ER stress markers in the lungs of allergic mice. Quantitative mRNA analysis of airway epithelial brushings from human subjects demonstrated that several ATF6α-related transcripts were significantly upregulated in patients with asthma compared with healthy subjects. Collectively, these results demonstrate that CBA-based therapy potently inhibits the allergen-induced UPR and allergic airway disease in mice via preferential binding of the canonical transducer of the UPR, ATF6α. These results potentially suggest a novel avenue to treat allergic asthma using select CBAs.
Collapse
Affiliation(s)
- Emily M Nakada
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Nirav R Bhakta
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, UCSF School of Medicine, San Francisco, California, USA
| | - Bethany R Korwin-Mihavics
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Amit Kumar
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Nicolas Chamberlain
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Sierra R Bruno
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - David G Chapman
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA.,Translational Airways Group, Discipline of Medical Science, University of Technology Sydney, Ultimo, Australia.,Woolcock Institute of Medical Research, University of Sydney, Glebe, Australia
| | - Sidra M Hoffman
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Nirav Daphtary
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Minara Aliyeva
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Charles G Irvin
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Anne E Dixon
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Prescott G Woodruff
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, UCSF School of Medicine, San Francisco, California, USA
| | - Shantu Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Matthew E Poynter
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Dhimant H Desai
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| |
Collapse
|
25
|
Bhakta NR, Christenson SA, Nerella S, Solberg OD, Nguyen CP, Choy DF, Jung KL, Garudadri S, Bonser LR, Pollack JL, Zlock LT, Erle DJ, Langelier C, Derisi JL, Arron JR, Fahy JV, Woodruff PG. IFN-stimulated Gene Expression, Type 2 Inflammation, and Endoplasmic Reticulum Stress in Asthma. Am J Respir Crit Care Med 2019; 197:313-324. [PMID: 29064281 DOI: 10.1164/rccm.201706-1070oc] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Quantification of type 2 inflammation provided a molecular basis for heterogeneity in asthma. Non-type 2 pathways that contribute to asthma pathogenesis are not well understood. OBJECTIVES To identify dysregulated pathways beyond type 2 inflammation. METHODS We applied RNA sequencing to airway epithelial brushings obtained from subjects with stable mild asthma not on corticosteroids (n = 19) and healthy control subjects (n = 16). Sequencing reads were mapped to human and viral genomes. In the same cohort, and in a separate group with severe asthma (n = 301), we profiled blood gene expression with microarrays. MEASUREMENTS AND MAIN RESULTS In airway brushings from mild asthma on inhaled corticosteroids, RNA sequencing yielded 1,379 differentially expressed genes (false discovery rate < 0.01). Pathway analysis revealed increased expression of type 2 markers, IFN-stimulated genes (ISGs), and endoplasmic reticulum (ER) stress-related genes. Airway epithelial ISG expression was not associated with type 2 inflammation in asthma or with viral transcripts but was associated with reduced lung function by FEV1 (ρ = -0.72; P = 0.0004). ER stress was confirmed by an increase in XBP1 (X-box binding protein 1) splicing in mild asthma and was associated with both type 2 inflammation and ISG expression. ISGs were also the most activated genes in blood cells in asthma and were correlated with airway ISG expression (ρ = 0.55; P = 0.030). High blood ISG expression in severe asthma was similarly unrelated to type 2 inflammation. CONCLUSIONS ISG activation is prominent in asthma, independent of viral transcripts, orthogonal to type 2 inflammation, and associated with distinct clinical features. ER stress is associated with both type 2 inflammation and ISG expression.
Collapse
Affiliation(s)
- Nirav R Bhakta
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | | | - Srilaxmi Nerella
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | - Owen D Solberg
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | - Christine P Nguyen
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | - David F Choy
- 2 Genentech, Inc., South San Francisco, California; and
| | - Kyle L Jung
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | - Suresh Garudadri
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine
| | - Luke R Bonser
- 3 Cardiovascular Research Institute.,4 Lung Biology Center, Department of Medicine
| | | | | | - David J Erle
- 3 Cardiovascular Research Institute.,4 Lung Biology Center, Department of Medicine
| | - Charles Langelier
- 6 Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Joseph L Derisi
- 7 Department of Biochemistry and Biophysics, University of California at San Francisco and Howard Hughes Medical Institute, San Francisco, California
| | | | - John V Fahy
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine.,3 Cardiovascular Research Institute
| | - Prescott G Woodruff
- 1 Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine.,3 Cardiovascular Research Institute
| |
Collapse
|
26
|
Fehl J, Pozarska A, Nardiello C, Rath P, Surate Solaligue DE, Vadász I, Mayer K, Herold S, Seeger W, Morty RE. Control Interventions Can Impact Alveolarization and the Transcriptome in Developing Mouse Lungs. Anat Rec (Hoboken) 2018; 302:346-363. [PMID: 30412359 DOI: 10.1002/ar.23931] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/23/2017] [Accepted: 02/02/2018] [Indexed: 01/20/2023]
Abstract
There is currently much interest in understanding the mechanisms of normal and aberrant lung alveolarization, particularly in the context of bronchopulmonary dysplasia, a common complication of preterm birth where alveolarization is impeded. To this end, the parenteral administration of pharmacological agents that modulate biochemical pathways, or facilitate modulation of gene expression in transgenic animals, has facilitated the discovery and validation of mechanisms that direct lung development. Such studies include control interventions, where the solvent vehicle, perhaps containing an inactive form of the agent applied, is administered; thereby providing a well-controlled point of reference for the analysis of the partner experiment. In the present study, the impact of several widely used control interventions in developing C57Bl/6J mouse pups was examined for effects on lung structure and the lung transcriptome. Parenteral administration of scrambled microRNA inhibitors (called antagomiRs) that are used to control in vivo microRNA neutralization studies, impacted lung volume, septal thickness, and the transcriptome of developing mouse lungs; with some effects dependent upon nucleotide sequence. Repeated intraperitoneal isotonic saline injections altered lung volume, with limited impact on the transcriptome. Parenteral administration of the tamoxifen solvent Miglyol accelerated mouse pup growth, and changed the abundance of 73 mRNA transcripts in the lung. Tamoxifen applied in Miglyol-in the absence of Cre recombinase-decreased pup growth, lung volume, and lung alveolarization and changed the abundance of 298 mRNA transcripts in the lung. These data demonstrate that widely used control interventions can directly impact lung alveolarization and the lung transcriptome in studies on lung development. Anat Rec, 302:346-363, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joshua Fehl
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Agnieszka Pozarska
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Philipp Rath
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Konstantin Mayer
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
27
|
Chen ACH, Burr L, McGuckin MA. Oxidative and endoplasmic reticulum stress in respiratory disease. Clin Transl Immunology 2018; 7:e1019. [PMID: 29928501 PMCID: PMC5999202 DOI: 10.1002/cti2.1019] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/01/2018] [Accepted: 05/03/2018] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress and endoplasmic reticulum (ER) stress are related states that can occur in cells as part of normal physiology but occur frequently in diseases involving inflammation. In this article, we review recent findings relating to the role of oxidative and ER stress in the pathophysiology of acute and chronic nonmalignant diseases of the lung, including infections, cystic fibrosis, idiopathic pulmonary fibrosis and asthma. We also explore the potential of drugs targeting oxidative and ER stress pathways to alleviate disease.
Collapse
Affiliation(s)
- Alice C-H Chen
- Diamantina Institute Faculty of Medicine The University of Queensland Brisbane QLD Australia.,Department of Cell and Molecular Therapy Royal Prince Alfred Hospital Sydney NSW Australia
| | - Lucy Burr
- Department of Respiratory Medicine Mater Adult Hospital and Mater Research Institute - The University of Queensland Raymond Tce, South Brisbane QLD Australia
| | - Michael A McGuckin
- Inflammatory Disease Biology and Therapeutics Group Translational Research Institute Mater Research Institute - The University of Queensland Brisbane QLD Australia
| |
Collapse
|
28
|
Pathinayake PS, Hsu ACY, Waters DW, Hansbro PM, Wood LG, Wark PAB. Understanding the Unfolded Protein Response in the Pathogenesis of Asthma. Front Immunol 2018; 9:175. [PMID: 29472925 PMCID: PMC5810258 DOI: 10.3389/fimmu.2018.00175] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/19/2018] [Indexed: 01/14/2023] Open
Abstract
Asthma is a heterogeneous, chronic inflammatory disease of the airways. It is a complex disease with different clinical phenotypes and results in a substantial socioeconomic burden globally. Poor understanding of pathogenic mechanisms of the disease hinders the investigation into novel therapeutics. Emerging evidence of the unfolded protein response (UPR) in the endoplasmic reticulum (ER) has demonstrated previously unknown functions of this response in asthma development. A worsening of asthmatic condition can be brought on by stimuli such as oxidative stress, pathogenic infections, and allergen exposure. All of which can induce ER stress and activate UPR leading to activation of different inflammatory responses and dysregulate the innate immune functions in the airways. The UPR as a central regulator of asthma pathogenesis may explain several unknown mechanism of the disease onset, which leads us in new directions for future asthma treatments. In this review, we summarize and discuss the causes and impact of ER–UPR in driving the pathogenesis of asthma and highlight its importance in clinical implications.
Collapse
Affiliation(s)
- Prabuddha S Pathinayake
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Alan C-Y Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - David W Waters
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
29
|
Rath P, Nardiello C, Morty RE. A new target for caffeine in the developing lung: endoplasmic reticulum stress? Am J Physiol Lung Cell Mol Physiol 2017; 313:L659-L663. [DOI: 10.1152/ajplung.00251.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 01/12/2023] Open
Affiliation(s)
- Philipp Rath
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and
| | - Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E. Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
30
|
Ghrelin Ameliorates Asthma by Inhibiting Endoplasmic Reticulum Stress. Am J Med Sci 2017; 354:617-625. [PMID: 29208260 DOI: 10.1016/j.amjms.2017.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND This study aimed to confirm the ameliorative effect of ghrelin on asthma and investigate its mechanism. MATERIALS AND METHODS The murine model of asthma was induced by ovalbumin (OVA) treatment and assessed by histological pathology and airway responsiveness to methacholine. The total and differential leukocytes were counted. Tumor necrosis factor α, interferon γ, interleukin-5 and interleukin-13 levels in bronchoalveolar lavage fluid were quantified by commercial kits. The protein levels in pulmonary tissues were measured by Western blot analysis. RESULTS Ghrelin ameliorated the histological pathology and airway hyperresponsiveness in the OVA-induced asthmatic mouse model. Consistently, OVA-increased total and differential leukocytes and levels of tumor necrosis factor α, interferon γ, interleukin-5 and interleukin-13 in bronchoalveolar lavage fluid were significantly attenuated by ghrelin. Ghrelin prevented the increased protein levels of the endoplasmic reticulum stress markers glucose regulated protein 78 and CCAAT/enhancer binding protein homologous protein and reversed the reduced levels of p-Akt in asthmatic mice. CONCLUSIONS Ghrelin might prevent endoplasmic reticulum stress activation by stimulating the Akt signaling pathway, which attenuated inflammation and ameliorated asthma in mice. Ghrelin might be a new target for asthma therapy.
Collapse
|
31
|
Modulation of the Unfolded Protein Response by Tauroursodeoxycholic Acid Counteracts Apoptotic Cell Death and Fibrosis in a Mouse Model for Secondary Biliary Liver Fibrosis. Int J Mol Sci 2017; 18:ijms18010214. [PMID: 28117681 PMCID: PMC5297843 DOI: 10.3390/ijms18010214] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 01/09/2017] [Accepted: 01/13/2017] [Indexed: 12/12/2022] Open
Abstract
The role of endoplasmic reticulum stress and the unfolded protein response (UPR) in cholestatic liver disease and fibrosis is not fully unraveled. Tauroursodeoxycholic acid (TUDCA), a hydrophilic bile acid, has been shown to reduce endoplasmic reticulum (ER) stress and counteract apoptosis in different pathologies. We aimed to investigate the therapeutic potential of TUDCA in experimental secondary biliary liver fibrosis in mice, induced by common bile duct ligation. The kinetics of the hepatic UPR and apoptosis during the development of biliary fibrosis was studied by measuring markers at six different timepoints post-surgery by qPCR and Western blot. Next, we investigated the therapeutic potential of TUDCA, 10 mg/kg/day in drinking water, on liver damage (AST/ALT levels) and fibrosis (Sirius red-staining), in both a preventive and therapeutic setting. Common bile duct ligation resulted in the increased protein expression of CCAAT/enhancer-binding protein homologous protein (CHOP) at all timepoints, along with upregulation of pro-apoptotic caspase 3 and 12, tumor necrosis factor receptor superfamily, member 1A (TNFRsf1a) and Fas-Associated protein with Death Domain (FADD) expression. Treatment with TUDCA led to a significant reduction of liver fibrosis, accompanied by a slight reduction of liver damage, decreased hepatic protein expression of CHOP and reduced gene and protein expression of pro-apoptotic markers. These data indicate that TUDCA exerts a beneficial effect on liver fibrosis in a model of cholestatic liver disease, and suggest that this effect might, at least in part, be attributed to decreased hepatic UPR signaling and apoptotic cell death.
Collapse
|
32
|
Prakash YS. Emerging concepts in smooth muscle contributions to airway structure and function: implications for health and disease. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1113-L1140. [PMID: 27742732 DOI: 10.1152/ajplung.00370.2016] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/06/2016] [Indexed: 12/15/2022] Open
Abstract
Airway structure and function are key aspects of normal lung development, growth, and aging, as well as of lung responses to the environment and the pathophysiology of important diseases such as asthma, chronic obstructive pulmonary disease, and fibrosis. In this regard, the contributions of airway smooth muscle (ASM) are both functional, in the context of airway contractility and relaxation, as well as synthetic, involving production and modulation of extracellular components, modulation of the local immune environment, cellular contribution to airway structure, and, finally, interactions with other airway cell types such as epithelium, fibroblasts, and nerves. These ASM contributions are now found to be critical in airway hyperresponsiveness and remodeling that occur in lung diseases. This review emphasizes established and recent discoveries that underline the central role of ASM and sets the stage for future research toward understanding how ASM plays a central role by being both upstream and downstream in the many interactive processes that determine airway structure and function in health and disease.
Collapse
Affiliation(s)
- Y S Prakash
- Departments of Anesthesiology, and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|