1
|
Chen R, Cui Y, Ip MSM, Mak JCW. Cigarette smoke induces endoplasmic reticulum stress-associated mucus hypersecretion via orosomucoid 1-like protein 3 in airway epithelia. Free Radic Res 2025:1-12. [PMID: 40317248 DOI: 10.1080/10715762.2025.2501019] [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: 05/17/2024] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Apart from a strong association with childhood-onset asthma, orosomucoid 1-like protein 3 (ORMDL3), an endoplasmic reticulum (ER)-localized transmembrane protein, is also linked with chronic obstructive pulmonary disease (COPD), in which cigarette smoke (CS) is the crucial risk factor. Compared to healthy subjects, COPD patients had elevated ORMDL3 mRNA in well-differentiated primary human bronchial epithelial cells (HBECs). However, its role in COPD remains understudied. We therefore hypothesize that ORMDL3 may play an essential role in CS-induced chronic mucus hypersecretion and inflammation via activation of specific unfolded protein response (UPR) pathways under ER stress in primary HBECs. Gene silencing using siRNA for ORMDL3 was performed in submerged culture of primary HBECs before 24-hour cigarette smoke medium (CSM) exposure. The mucin, inflammatory and mitochondrial markers, and the activation of the UPR pathways were evaluated. CSM triggered significant induction of ORMDL3 expression at both mRNA and protein level, which was significantly inhibited by silencing ORMDL3. In addition, ORMDL3 knockdown inhibited CSM-induced mucin MUC5AC mRNA and release of inflammatory marker interleukin (IL)-8. Silencing ORMDL3 reduced CSM-induced ER stress via inhibiting the activating transcription factor (ATF)6 and the inositol-requiring enzyme (IRE)1 of the UPR pathways. The involvement of ORMDL3 was demonstrated in mitochondrial dynamics via fusion protein Mfn2 and mitochondrial respiration after CSM stimulation. In conclusion, ORMDL3 is an inducible gene in mediating CS-induced activation of specific ATF6 and IRE1 pathways to regulate mucus hypersecretion and inflammation. Therefore, ORMDL3 may be a promising therapeutic target to treat smoking-associated mucus hypersecretion and inflammation in COPD.
Collapse
Affiliation(s)
- Rui Chen
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology and Infection, Hong Kong Science Park, Hong Kong SAR, China
| | - Yuting Cui
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Mary Sau-Man Ip
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Judith Choi-Wo Mak
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
2
|
Sugiura S, Yoshida H, Sugiura H, Uehara M, Sugiura Y, Maruo Y, Hayashi Y, Yamamoto T, Kato T, Fujimoto N, Udagawa J. Increased intracellular stress responses and decreased KLF2 in adult patients with atopic dermatitis. Cell Stress Chaperones 2025; 30:84-99. [PMID: 39938773 PMCID: PMC11891603 DOI: 10.1016/j.cstres.2025.02.001] [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: 09/08/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
Atopic dermatitis (AD) is prone to exacerbations in response to various triggering factors and flare-ups after remission. We searched for molecules associated with relapse/exacerbation of AD among molecules with altered gene expression in the skin of patients with AD. Microarray analyses were performed on lesional and nonlesional skin of adolescent or adult patients with recalcitrant AD and healthy controls. Five chaperones involved in intracellular stress responses, namely heat shock protein family A (Hsp70) member 9 (HSPA9), heat shock protein 90 beta family member 1 (HSP90B1), calnexin (CANX), malectin (MLEC; endoplasmic reticulum-associated degradation), and heat shock protein family D (Hsp60) member 1 (HSPD1), were consistently upregulated in involved and uninvolved skin of patients with AD. Damage-associated molecular patterns were upregulated in involved skin. KLF transcription factor 2 (KLF2) was decreased in involved skin and exhibited a decreasing trend in uninvolved skin of patients with AD. CD4(+)/CD8(+) double-positive cells (1.4% of T cells) were detected in lesions with declined KLF2 levels. WNT inhibitory factor 1 (WIF1) was downregulated in involved skin. Prolactin-induced protein was upregulated in only uninvolved skin of patients with AD. We found increased intracellular stress responses and decreased expression of KLF2 in the skin of patients with AD. Multifactorial genetic diseases, such as asthma, inflammatory bowel disease, type 2 diabetes, and rheumatoid arthritis, are associated with intracellular stress. Intracellular abnormalities may also be responsible for AD. Further research on AD may incorporate enhanced intracellular stress response and the decreased expression of KLF2 into the mechanism underlying AD.
Collapse
Affiliation(s)
- Shuji Sugiura
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan; Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan.
| | - Hiderou Yoshida
- Department of Molecular Biochemistry, Graduate School of Life Science, University of Hyogo, Ako, Japan
| | - Hisashi Sugiura
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan; Department of Dermatology, Sugiura Dermatology Clinic, Kusatsu, Japan
| | - Masami Uehara
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Yasuo Sugiura
- International Health Care Center, National Center for Global Health and Medicine, Tokyo, Japan; Department of Pediatrics, Navitas Clinic, Tokyo, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Yuji Hayashi
- Hospital Division of Diagnostic Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Takefumi Yamamoto
- Central Research Laboratory, Shiga University of Medical Science, Otsu, Japan
| | - Takeshi Kato
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Noriki Fujimoto
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Jun Udagawa
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
3
|
Fu Z, Wang W, Gao Y. Understanding the impact of ER stress on lung physiology. Front Cell Dev Biol 2024; 12:1466997. [PMID: 39744015 PMCID: PMC11688383 DOI: 10.3389/fcell.2024.1466997] [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: 07/24/2024] [Accepted: 11/22/2024] [Indexed: 01/04/2025] Open
Abstract
Human lungs consist of a distinctive array of cell types, which are subjected to persistent challenges from chemical, mechanical, biological, immunological, and xenobiotic stress throughout life. The disruption of endoplasmic reticulum (ER) homeostatic function, triggered by various factors, can induce ER stress. To overcome the elevated ER stress, an adaptive mechanism known as the unfolded protein response (UPR) is activated in cells. However, persistent ER stress and maladaptive UPR can lead to defects in proteostasis at the cellular level and are typical features of the lung aging. The aging lung and associated lung diseases exhibit signs of ER stress-related disruption in cellular homeostasis. Dysfunction resulting from ER stress and maladaptive UPR can compromise various cellular and molecular processes associated with aging. Hence, comprehending the mechanisms of ER stress and UPR components implicated in aging and associated lung diseases could enable to develop appropriate therapeutic strategies for the vulnerable population.
Collapse
Affiliation(s)
- Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wei Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuan Gao
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
4
|
Wang H, Zhang L, Shang Y. DEPTOR attenuates asthma progression by suppressing endoplasmic reticulum stress through SOD1. Biol Direct 2024; 19:114. [PMID: 39533404 PMCID: PMC11556204 DOI: 10.1186/s13062-024-00557-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Endoplasmic reticulum (ER) stress has been shown to play a pivotal role in the pathogenesis of asthma. DEPTOR (DEP Domain Containing MTOR Interacting Protein) is an endogenous mTOR inhibitor that participates in various physiological processes such as cell growth, apoptosis, autophagy, and ER homeostasis. However, the role of DEPTOR in the pathogenesis of asthma is still unknown. In this study, an ovalbumin (OVA)-induced mice model and IL-13 induced 16HBE cells were used to evaluate the effect of DEPTOR on asthma. A decreased DEPTOR expression was shown in the lung tissues of OVA-mice and IL-13 induced 16HBE cells. Upregulation of DEPTOR attenuated airway goblet cell hyperplasia, inhibited mucus hypersecretion, decreased the expression of mucin MUC5AC, and suppressed the level of inflammatory factors IL-4 and IL-5, which were all induced by OVA treatment. The increased protein expression of ER stress markers GRP78, CHOP, unfolded protein response (UPR) related proteins, and apoptosis markers in OVA mice were also inhibited by DEPTOR overexpression. In IL-13 induced 16HBE cells, overexpression of DEPTOR decreased IL-4, IL-5, and MUC5AC levels, preventing ER stress response and UPR process. Furthermore, from the proteomics results, we identified that SOD1 (Cu/Zn Superoxide Dismutase 1) may be the downstream factor of DEPTOR. Similar to DEPTOR, upregulation of SOD1 alleviated asthma progression. Rescue experiments showed that SOD1 inhibition abrogates the remission effect of DEPTOR on ER stress in vitro. In conclusion, these data suggested that DEPTOR attenuates asthma progression by suppressing endoplasmic reticulum stress through SOD1.
Collapse
Affiliation(s)
- Hao Wang
- Department of Pediatric Respiratory Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Lei Zhang
- Department of General Surgery, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Yunxiao Shang
- Department of Pediatric Respiratory Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China.
| |
Collapse
|
5
|
Zhang W, Zhang C, Zhang Y, Zhou X, Dong B, Tan H, Su H, Sun X. Multifaceted roles of mitochondria in asthma. Cell Biol Toxicol 2024; 40:85. [PMID: 39382744 PMCID: PMC11464602 DOI: 10.1007/s10565-024-09928-8] [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: 06/10/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024]
Abstract
Mitochondria are essential organelles within cells, playing various roles in numerous cellular processes, including differentiation, growth, apoptosis, energy conversion, metabolism, and cellular immunity. The phenotypic variation of mitochondria is specific to different tissues and cell types, resulting in significant differences in their function, morphology, and molecular characteristics. Asthma is a chronic, complex, and heterogeneous airway disease influenced by external factors such as environmental pollutants and allergen exposure, as well as internal factors at the tissue, cellular, and genetic levels, including lung and airway structural cells, immune cells, granulocytes, and mast cells. Therefore, a comprehensive understanding of the specific responses of mitochondria to various external environmental stimuli and internal changes are crucial for elucidating the pathogenesis of asthma. Previous research on mitochondrial-targeted therapy for asthma has primarily focused on antioxidants. Consequently, it is necessary to summarize the multifaceted roles of mitochondria in the pathogenesis of asthma to discover additional strategies targeting mitochondria in this context. In this review, our goal is to describe the changes in mitochondrial function in response to various exposure factors across different cell types and other relevant factors in the context of asthma, utilizing a new mitochondrial terminology framework that encompasses cell-dependent mitochondrial characteristics, molecular features, mitochondrial activity, function, and behavior.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Chenyu Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yi Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xuehua Zhou
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bo Dong
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hong Tan
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
6
|
Pathinayake PS, Hsu ACY, Nichol KS, Horvat JC, Hansbro PM, Wark PAB. Endoplasmic reticulum stress enhances the expression of TLR3-induced TSLP by airway epithelium. Am J Physiol Lung Cell Mol Physiol 2024; 326:L618-L626. [PMID: 38469627 PMCID: PMC11381004 DOI: 10.1152/ajplung.00378.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 03/13/2024] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is an epithelial-derived pleiotropic cytokine that regulates T-helper 2 (Th2) immune responses in the lung and plays a major role in severe uncontrolled asthma. Emerging evidence suggests a role for endoplasmic reticulum (ER) stress in the pathogenesis of asthma. In this study, we determined if ER stress and the unfolded protein response (UPR) signaling are involved in TSLP induction in the airway epithelium. For this, we treated human bronchial epithelial basal cells and differentiated primary bronchial epithelial cells with ER stress inducers and the TSLP mRNA and protein expression was determined. A series of siRNA gene knockdown experiments were conducted to determine the ER stress-induced TSLP signaling pathways. cDNA collected from asthmatic bronchial biopsies was used to determine the gene correlation between ER stress and TSLP. Our results show that ER stress signaling induces TSLP mRNA expression via the PERK-C/EBP homologous protein (CHOP) signaling pathway. AP-1 transcription factor is important in regulating this ER stress-induced TSLP mRNA induction, though ER stress alone cannot induce TSLP protein production. However, ER stress significantly enhances TLR3-induced TSLP protein secretion in the airway epithelium. TSLP and ER stress (PERK) mRNA expression positively correlates in bronchial biopsies from participants with asthma, particularly in neutrophilic asthma. In conclusion, these results suggest that ER stress primes TSLP that is then enhanced further upon TLR3 activation, which may induce severe asthma exacerbations. Targeting ER stress using pharmacological interventions may provide novel therapeutics for severe uncontrolled asthma.NEW & NOTEWORTHY TSLP is an epithelial-derived cytokine and a key regulator in the pathogenesis of severe uncontrolled asthma. We demonstrate a novel mechanism by which endoplasmic reticulum stress signaling upregulates airway epithelial TSLP mRNA expression via the PERK-CHOP signaling pathway and enhances TLR3-mediated TSLP protein secretion.
Collapse
Affiliation(s)
- Prabuddha S Pathinayake
- Immune Health Program, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Alan C-Y Hsu
- Immune Health Program, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
- Signature Research Program in Emerging Infectious Diseases, Duke-National University of Singapore (NUS) Graduate Medical School, Singapore, Singapore
| | - Kristy S Nichol
- Immune Health Program, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Immune Health Program, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Immune Health Program, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Peter A B Wark
- Immune Health Program, Hunter Medical Research Institute and School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
- School of Medicine, Monash University, Melbourne, Victoria, Australia
- AIRMED Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Leonardi A, Donato A, Rosani U, Di Stefano A, Cavarzeran F, Brun P. Endoplasmic Reticulum Stress and Unfolded Protein Response in Vernal Keratoconjunctivitis. Invest Ophthalmol Vis Sci 2024; 65:23. [PMID: 38597723 PMCID: PMC11008754 DOI: 10.1167/iovs.65.4.23] [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: 02/06/2024] [Accepted: 03/20/2024] [Indexed: 04/11/2024] Open
Abstract
Purpose Vernal keratoconjunctivitis (VKC) is an ocular allergic disease characterized by a type 2 inflammation, tissue remodeling, and low quality of life for the affected patients. We investigated the involvement of endoplasmic reticulum (ER) stress and unfolded protein response in VKC. Methods Conjunctival imprints from VKC patients and normal subjects (CTs) were collected, and RNA was isolated, reverse transcribed, and analyzed with the Affymetrix microarray. Differentially expressed genes between VKC patients and CTs were evaluated. Genes related to ER stress, apoptosis, and autophagy were further considered. VKC and CT conjunctival biopsies were analyzed by immunohistochemistry (IHC) with specific antibodies against unfolded protein response (UPR), apoptosis, and inflammation. Conjunctival fibroblast and epithelial cell cultures were exposed to the conditioned medium of activated U937 monocytes and analyzed by quantitative PCR for the expression of UPR, apoptosis, autophagy, and inflammatory markers. Results ER chaperones HSPA5 (GRP78/BiP) and HYOU1 (GRP170) were upregulated in VKC patients compared to CTs. Genes encoding for ER transmembrane proteins, PKR-like ER kinase (PERK), activating transcription factor 6 (ATF6), ER-associated degradation (ERAD), and autophagy were upregulated, but not those related to apoptosis. Increased positive reactivity of BiP and ATF6 and unchanged expression of apoptosis markers were confirmed by IHC. Cell cultures in stress conditions showed an overexpression of UPR, proinflammatory, apoptosis, and autophagy markers. Conclusions A significant overexpression of genes encoding for ER stress, UPR, and pro-inflammatory pathway components was reported for VKC. Even though these pathways may lead to ER homeostasis, apoptosis, or inflammation, ER stress in VKC may predominantly contribute to promote inflammation.
Collapse
Affiliation(s)
- Andrea Leonardi
- Ophthalmology Unit, Department of Neuroscience, University of Padova, Padova, Italy
| | - Alice Donato
- Histology Unit, Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Antonino Di Stefano
- Istituti Clinici Scientifici Maugeri, IRCCS, Istituto Scientifico di Veruno, Veruno, Italy
| | - Fabiano Cavarzeran
- Ophthalmology Unit, Department of Neuroscience, University of Padova, Padova, Italy
| | - Paola Brun
- Histology Unit, Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
8
|
Koller A, Filosi M, Weissensteiner H, Fazzini F, Gorski M, Pattaro C, Schönherr S, Forer L, Herold JM, Stark KJ, Döttelmayer P, Hicks AA, Pramstaller PP, Würzner R, Eckardt KU, Heid IM, Fuchsberger C, Lamina C, Kronenberg F. Nuclear and mitochondrial genetic variants associated with mitochondrial DNA copy number. Sci Rep 2024; 14:2083. [PMID: 38267512 PMCID: PMC10808213 DOI: 10.1038/s41598-024-52373-0] [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: 08/29/2023] [Accepted: 01/17/2024] [Indexed: 01/26/2024] Open
Abstract
Mitochondrial DNA copy number (mtDNA-CN) is a biomarker for mitochondrial dysfunction associated with several diseases. Previous genome-wide association studies (GWAS) have been performed to unravel underlying mechanisms of mtDNA-CN regulation. However, the identified gene regions explain only a small fraction of mtDNA-CN variability. Most of this data has been estimated from microarrays based on various pipelines. In the present study we aimed to (1) identify genetic loci for qPCR-measured mtDNA-CN from three studies (16,130 participants) using GWAS, (2) identify potential systematic differences between our qPCR derived mtDNA-CN measurements compared to the published microarray intensity-based estimates, and (3) disentangle the nuclear from mitochondrial regulation of the mtDNA-CN phenotype. We identified two genome-wide significant autosomal loci associated with qPCR-measured mtDNA-CN: at HBS1L (rs4895440, p = 3.39 × 10-13) and GSDMA (rs56030650, p = 4.85 × 10-08) genes. Moreover, 113/115 of the previously published SNPs identified by microarray-based analyses were significantly equivalent with our findings. In our study, the mitochondrial genome itself contributed only marginally to mtDNA-CN regulation as we only detected a single rare mitochondrial variant associated with mtDNA-CN. Furthermore, we incorporated mitochondrial haplogroups into our analyses to explore their potential impact on mtDNA-CN. However, our findings indicate that they do not exert any significant influence on our results.
Collapse
Affiliation(s)
- Adriana Koller
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, 6020, Innsbruck, Austria
| | - Michele Filosi
- Eurac Research, Institute for Biomedicine, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Hansi Weissensteiner
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, 6020, Innsbruck, Austria
| | - Federica Fazzini
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, 6020, Innsbruck, Austria
| | - Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, 6020, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, 6020, Innsbruck, Austria
| | - Janina M Herold
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Klaus J Stark
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Patricia Döttelmayer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, 6020, Innsbruck, Austria
| | - Andrew A Hicks
- Eurac Research, Institute for Biomedicine, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Peter P Pramstaller
- Eurac Research, Institute for Biomedicine, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Reinhard Würzner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- German Chronic Kidney Disease Study, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Christian Fuchsberger
- Eurac Research, Institute for Biomedicine, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, 6020, Innsbruck, Austria
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstrasse 41, 6020, Innsbruck, Austria.
| |
Collapse
|
9
|
Beilankouhi EAV, Sajadi MA, Alipourfard I, Hassani P, Valilo M, Safaralizadeh R. Role of the ER-induced UPR pathway, apoptosis, and autophagy in colorectal cancer. Pathol Res Pract 2023; 248:154706. [PMID: 37499516 DOI: 10.1016/j.prp.2023.154706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
When large amounts of misfolded or unfolded proteins accumulate in the endoplasmic reticulum (ER) in response to stress, a process called unfolded protein response (UPR) is activated. The disruption of this process leads to many diseases including diabetes, neurodegenerative diseases, and many cancers. In the process of UPR in response to stress and unfolded proteins, specific signaling pathways are induced in the endoplasmic reticulum and subsequently transmitted to the nucleus and cytoplasm, causing homeostasis and restoring the cell's normal condition with reducing protein translation and synthesis. The UPR response followed by stress enhancement balances cell survival with death, therefore in this condition cells decide either to survive or have the path of apoptosis ahead. However, in some cases, this balance is disturbed and the UPR pathway is chronically activated or not activated and the cell conditions lead to cancer. This study aimed to briefly investigate the association between ER stress, UPR, apoptosis, and autophagy in colorectal cancer (CRC). Moreover, in current study, we will try to demonstrate canonical ways and methods for the treatment of CRC cells with attenuated ER stress.
Collapse
Affiliation(s)
| | | | - Iraj Alipourfard
- Insttue of Biology, Biotechnology and Environmental Protection, Faculty of Natural Science, University of Silesia, Katowice, Poland
| | - Peyman Hassani
- DVM Graduated, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
10
|
Wu D, Zhang X, Zimmerly KM, Wang R, Wang C, Hunter R, Wu X, Campen M, Liu M, Yang XO. Unfolded protein response factor ATF6 augments T helper cell responses and promotes mixed granulocytic airway inflammation. Mucosal Immunol 2023; 16:499-512. [PMID: 37209959 PMCID: PMC10530451 DOI: 10.1016/j.mucimm.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
The unfolded protein response (UPR) is associated with the risk of asthma, including treatment-refractory severe asthma. Recent studies demonstrated a pathogenic role of activating transcription factor 6a (ATF6a or ATF6), an essential UPR sensor, in airway structural cells. However, its role in T helper (TH) cells has not been well examined. In this study, we found that ATF6 was selectively induced by signal transducer and activator of transcription6 (STAT6) and STAT3 in TH2 and TH17 cells, respectively. ATF6 upregulated UPR genes and promoted the differentiation and cytokine secretion of TH2 and TH17 cells. T cell-specific Atf6-deficiency impaired TH2 and TH17 responses in vitro and in vivo and attenuated mixed granulocytic experimental asthma. ATF6 inhibitor Ceapin A7 suppressed the expression of ATF6 downstream genes and TH cell cytokines by both murine and human memory clusters of differentiation 4 (CD4)+ T cells. At the chronic stage of asthma, administration of Ceapin A7 lessened TH2 and TH17 responses, leading to alleviation of both airway neutrophilia and eosinophilia. Thus, our results demonstrate a critical role of ATF6 in TH2 and TH17 cell-driven mixed granulocytic airway disease, suggesting a novel option to combat steroid-resistant mixed and even T2-low endotypes of asthma by targeting ATF6.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Kourtney M Zimmerly
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Chunqing Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Russell Hunter
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA; Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Matthew Campen
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA.
| | - Xuexian O Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA.
| |
Collapse
|
11
|
Brasier AR. Innate Immunity, Epithelial Plasticity, and Remodeling in Asthma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:265-285. [PMID: 37464126 DOI: 10.1007/978-3-031-32259-4_13] [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: 07/20/2023]
Abstract
Innate immune responses (IIR) of the epithelium play a critical role in the initiation and progression of asthma. The core of the IIR is an intracellular signaling pathway activated by pattern recognition receptors (PRRs) to limit the spread of infectious organisms. This chapter will focus on the epithelium as the major innate sentinel cell and its role in acute exacerbations (AEs). Although the pathways of how the IIR activates the NFκB transcription factor, triggering cytokine secretion, dendritic cell activation, and Th2 polarization are well-described, recent exciting work has developed mechanistic insights into how chronic activation of the IIR is linked to mucosal adaptive responses. These adaptations include changes in cell state, now called epithelial-mesenchymal plasticity (EMP). EMP is a coordinated, genomic response to airway injury disrupting epithelial barrier function, expanding the basal lamina, and producing airway remodeling. EMP is driven by activation of the unfolded protein response (UPR), a transcriptional response producing metabolic shunting of glucose through the hexosamine biosynthetic pathway (HBP) to protein N-glycosylation. NFκB signaling and UPR activation pathways potentiate each other in remodeling the basement membrane. Understanding of injury-repair process of epithelium provides new therapeutic targets for precision approaches to the treatment of asthma exacerbations and their sequelae.
Collapse
Affiliation(s)
- Allan R Brasier
- Department of Medicine and Institute for Clinical and Translational Research (ICTR), School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
12
|
Zhu T, Brown AP, Cai LP, Quon G, Ji H. Single-Cell RNA-Seq Analysis Reveals Lung Epithelial Cell Type-Specific Responses to HDM and Regulation by Tet1. Genes (Basel) 2022; 13:genes13050880. [PMID: 35627266 PMCID: PMC9140484 DOI: 10.3390/genes13050880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Tet1 protects against house dust mite (HDM)-induced lung inflammation in mice and alters the lung methylome and transcriptome. In order to explore the role of Tet1 in individual lung epithelial cell types in HDM-induced inflammation, we established a model of HDM-induced lung inflammation in Tet1 knockout and littermate wild-type mice, then studied EpCAM+ lung epithelial cells using single-cell RNA-seq analysis. We identified eight EpCAM+ lung epithelial cell types, among which AT2 cells were the most abundant. HDM challenge altered the relative abundance of epithelial cell types and resulted in cell type-specific transcriptomic changes. Bulk and cell type-specific analysis also showed that loss of Tet1 led to the altered expression of genes linked to augmented HDM-induced lung inflammation, including alarms, detoxification enzymes, oxidative stress response genes, and tissue repair genes. The transcriptomic regulation was accompanied by alterations in TF activities. Trajectory analysis supports that HDM may enhance the differentiation of AP and BAS cells into AT2 cells, independent of Tet1. Collectively, our data showed that lung epithelial cells had common and unique transcriptomic signatures of allergic lung inflammation. Tet1 deletion altered transcriptomic networks in various lung epithelial cells, which may promote allergen-induced lung inflammation.
Collapse
Affiliation(s)
- Tao Zhu
- California National Primate Research Center, University of California, Davis, CA 95616, USA; (T.Z.); (A.P.B.); (L.P.C.)
| | - Anthony P. Brown
- California National Primate Research Center, University of California, Davis, CA 95616, USA; (T.Z.); (A.P.B.); (L.P.C.)
| | - Lucy P. Cai
- California National Primate Research Center, University of California, Davis, CA 95616, USA; (T.Z.); (A.P.B.); (L.P.C.)
| | - Gerald Quon
- Department of Molecular and Cellular Biology, Genome Center, University of California, Davis, CA 95616, USA;
| | - Hong Ji
- California National Primate Research Center, University of California, Davis, CA 95616, USA; (T.Z.); (A.P.B.); (L.P.C.)
- Department of Anatomy, Physiology and Cell biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
- Correspondence: ; Tel.: +1-530-754-0679
| |
Collapse
|
13
|
Portelli MA, Rakkar K, Hu S, Guo Y, Adcock IM, Sayers I. Translational Analysis of Moderate to Severe Asthma GWAS Signals Into Candidate Causal Genes and Their Functional, Tissue-Dependent and Disease-Related Associations. FRONTIERS IN ALLERGY 2022; 2:738741. [PMID: 35386986 PMCID: PMC8974692 DOI: 10.3389/falgy.2021.738741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Asthma affects more than 300 million people globally and is both under diagnosed and under treated. The most recent and largest genome-wide association study investigating moderate to severe asthma to date was carried out in 2019 and identified 25 independent signals. However, as new and in-depth downstream databases become available, the translational analysis of these signals into target genes and pathways is timely. In this study, unique (U-BIOPRED) and publicly available datasets (HaploReg, Open Target Genetics and GTEx) were investigated for the 25 GWAS signals to identify 37 candidate causal genes. Additional traits associated with these signals were identified through PheWAS using the UK Biobank resource, with asthma and eosinophilic traits amongst the strongest associated. Gene expression omnibus dataset examination identified 13 candidate genes with altered expression profiles in the airways and blood of asthmatic subjects, including MUC5AC and STAT6. Gene expression analysis through publicly available datasets highlighted lung tissue cell specific expression, with both MUC5AC and SLC22A4 genes showing enriched expression in ciliated cells. Gene enrichment pathway and interaction analysis highlighted the dominance of the HLA-DQA1/A2/B1/B2 gene cluster across many immunological diseases including asthma, type I diabetes, and rheumatoid arthritis. Interaction and prediction analyses found IL33 and IL18R1 to be key co-localization partners for other genes, predicted that CD274 forms co-expression relationships with 13 other genes, including the HLA-DQA1/A2/B1/B2 gene cluster and that MUC5AC and IL37 are co-expressed. Drug interaction analysis revealed that 11 of the candidate genes have an interaction with available therapeutics. This study provides significant insight into these GWAS signals in the context of cell expression, function, and disease relationship with the view of informing future research and drug development efforts for moderate-severe asthma.
Collapse
Affiliation(s)
- Michael A Portelli
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, National Institute for Health Research Nottingham Biomedical Research Centre, Nottingham University Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Kamini Rakkar
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, National Institute for Health Research Nottingham Biomedical Research Centre, Nottingham University Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Sile Hu
- Data Science Institute, Imperial College London, London, United Kingdom
| | - Yike Guo
- Data Science Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- The National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ian Sayers
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, National Institute for Health Research Nottingham Biomedical Research Centre, Nottingham University Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
14
|
Chen R, Michaeloudes C, Liang Y, Bhavsar PK, Chung KF, Ip MSM, Mak JCW. ORMDL3 regulates cigarette smoke-induced endoplasmic reticulum stress in airway smooth muscle cells. J Allergy Clin Immunol 2022; 149:1445-1457.e5. [PMID: 34624393 DOI: 10.1016/j.jaci.2021.09.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Orosomucoid 1-like protein 3 (ORMDL3), a transmembrane protein localized in the endoplasmic reticulum (ER), has been genetically associated with chronic obstructive pulmonary disease (COPD), in addition to childhood-onset asthma. However, the functional role of ORMDL3 in the pathogenesis of COPD is still unknown. OBJECTIVE Because cigarette smoke is the major risk factor for COPD, we aimed to investigate the role of ORMDL3 in cigarette smoke-induced human airway smooth muscle cell (HASMC) injury. METHODS The mRNA and protein expression of ORMDL3 was examined in HASMCs from nonsmokers and smokers without or with COPD. Knockdown of ORMDL3 in primary healthy HASMCs was performed using small interfering RNA before exposure to cigarette smoke medium (CSM) for 24 hours. Inflammatory, proliferative/apoptotic, ER stress, and mitochondrial markers were evaluated. RESULTS Elevation of ORMDL3 mRNA and protein expression was observed in HASMCs of smokers without or with COPD. CSM caused significant upregulation of ORMDL3 expression in healthy nonsmokers. ORMDL3 knockdown regulated CSM-induced inflammation, cell proliferation, and apoptosis. Silencing ORMDL3 led to reduction of CSM-induced ER stress via inhibition of unfolded protein response pathways such as activating transcription factor 6 and protein kinase RNA-like ER kinase. ORMDL3 was also involved in CSM-induced mitochondrial dysfunction via the mitochondrial fission process. CONCLUSIONS We report the induction of ORMDL3 in HASMCs after cigarette smoke exposure. ORMDL3 may mediate cigarette smoke-induced activation of unfolded protein response pathways during airway smooth muscle cell injury.
Collapse
Affiliation(s)
- Rui Chen
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Charalambos Michaeloudes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Yingmin Liang
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Mary S M Ip
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Judith C W Mak
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China; Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
15
|
Perotin JM, Wheway G, Tariq K, Azim A, Ridley RA, Ward JA, Schofield JP, Barber C, Howarth P, Davies DE, Djukanovic R. Vulnerability to acid reflux of the airway epithelium in severe asthma. Eur Respir J 2022; 60:13993003.01634-2021. [PMID: 34996831 DOI: 10.1183/13993003.01634-2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 12/10/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Severe asthma is associated with multiple co-morbidities, including gastro-oesophageal reflux disease (GORD) which can contribute to exacerbation frequency and poor quality of life. Since epithelial dysfunction is an important feature in asthma, we hypothesised that in severe asthma the bronchial epithelium is more susceptible to the effects of acid reflux. METHODS We developed an in vitro model of GORD using differentiated bronchial epithelial cells (BECs) from normal or severe asthmatic donors exposed to a combination of pepsin, acid pH, and bile acids using a multiple challenge protocol (MCP-PAB). We also analysed bronchial biopsies and undertook RNA-sequencing of bronchial brushings from controls and severe asthmatics without or with GORD. RESULTS Exposure of BECs to the MCP-PAB caused structural disruption, increased permeability, IL-33 expression, inflammatory mediator release and changes in gene expression for multiple biological processes. Cultures from severe asthmatics were significantly more affected than those from healthy donors. Analysis of bronchial biopsies confirmed increased IL-33 expression in severe asthmatics with GORD. RNA-sequencing of bronchial brushings from this group identified 15 of the top 37 dysregulated genes found in MCP-PAB treated BECs, including genes involved in oxidative stress responses. CONCLUSIONS By affecting epithelial permeability, GORD may increase exposure of the airway submucosa to allergens and pathogens, resulting in increased risk of inflammation and exacerbations. CLINICAL IMPLICATION These results suggest the need for research into alternative therapeutic management of GORD in severe asthma.
Collapse
Affiliation(s)
- Jeanne-Marie Perotin
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK .,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Department of Respiratory Diseases, UMRS1250, University Hospital of Reims, France
| | - Gabrielle Wheway
- Human Development and Health, University of Southampton Faculty of Medicine, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Kamran Tariq
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Adnan Azim
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Robert A Ridley
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jonathan A Ward
- The Histochemistry Research Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - James Pr Schofield
- Centre for Proteomic Research, Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Clair Barber
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Peter Howarth
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Donna E Davies
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK.,joint senior authors
| | - Ratko Djukanovic
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, UK.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK.,joint senior authors
| |
Collapse
|
16
|
Resano A, Bhattacharjee S, Barajas M, Do KV, Aguado-Jiménez R, Rodríguez D, Palacios R, Bazán NG. Elovanoids Counteract Inflammatory Signaling, Autophagy, Endoplasmic Reticulum Stress, and Senescence Gene Programming in Human Nasal Epithelial Cells Exposed to Allergens. Pharmaceutics 2022; 14:113. [PMID: 35057008 PMCID: PMC8778361 DOI: 10.3390/pharmaceutics14010113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/13/2021] [Accepted: 12/25/2021] [Indexed: 11/28/2022] Open
Abstract
To contribute to further understanding the cellular and molecular complexities of inflammatory-immune responses in allergic disorders, we have tested the pro-homeostatic elovanoids (ELV) in human nasal epithelial cells (HNEpC) in culture challenged by several allergens. ELV are novel bioactive lipid mediators synthesized from the omega-3 very-long-chain polyunsaturated fatty acids (VLC-PUFA,n-3). We ask if: (a) several critical signaling events that sustain the integrity of the human nasal epithelium and other organ barriers are perturbed by house dust mites (HDM) and other allergens, and (b) if ELV would participate in beneficially modulating these events. HDM is a prevalent indoor allergen that frequently causes allergic respiratory diseases, including allergic rhinitis and allergic asthma, in HDM-sensitized individuals. Our study used HNEpC as an in vitro model to study the effects of ELV in counteracting HDM sensitization resulting in inflammation, endoplasmic reticulum (ER) stress, autophagy, and senescence. HNEpC were challenged with the following allergy inducers: LPS, poly(I:C), or Dermatophagoides farinae plus Dermatophagoides pteronyssinus extract (HDM) (30 µg/mL), with either phosphate-buffered saline (PBS) (vehicle) or ELVN-34 (500 nM). Results show that ELVN-34 promotes cell viability and reduces cytotoxicity upon HDM sensitization of HNEpC. This lipid mediator remarkably reduces the abundance of pro-inflammatory cytokines and chemokines IL-1β, IL-8, VEGF, IL-6, CXCL1, CCL2, and cell adhesion molecule ICAM1 and restores the levels of the pleiotropic anti-inflammatory IL-10. ELVN-34 also lessens the expression of senescence gene programming as well as of gene transcription engaged in pro-inflammatory responses. Our data also uncovered that HDM triggered the expression of key genes that drive autophagy, unfolded protein response (UPR), and matrix metalloproteinases (MMP). ELVN-34 has been shown to counteract these effects effectively. Together, our data reveal a novel, pro-homeostatic, cell-protective lipid-signaling mechanism in HNEpC as potential therapeutic targets for allergies.
Collapse
Affiliation(s)
- Alfredo Resano
- Department of Health Science, Public University of Navarra, 31006 Pamplona, Spain;
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, Louisiana State University Health New Orleans (LSUHSC), New Orleans, LA 70112, USA; (S.B.); (K.V.D.)
| | - Miguel Barajas
- Department of Health Science, Public University of Navarra, 31006 Pamplona, Spain;
| | - Khanh V. Do
- Neuroscience Center of Excellence, Louisiana State University Health New Orleans (LSUHSC), New Orleans, LA 70112, USA; (S.B.); (K.V.D.)
| | | | | | | | - Nicolás G. Bazán
- Neuroscience Center of Excellence, Louisiana State University Health New Orleans (LSUHSC), New Orleans, LA 70112, USA; (S.B.); (K.V.D.)
| |
Collapse
|
17
|
Weingartner M, Stücheli S, Jebbawi F, Gottstein B, Beldi G, Lundström-Stadelmann B, Wang J, Odermatt A. Albendazole reduces hepatic inflammation and endoplasmic reticulum-stress in a mouse model of chronic Echinococcus multilocularis infection. PLoS Negl Trop Dis 2022; 16:e0009192. [PMID: 35030165 PMCID: PMC8794265 DOI: 10.1371/journal.pntd.0009192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 01/27/2022] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Echinococcus multilocularis causes alveolar echinococcosis (AE), a rising zoonotic disease in the northern hemisphere. Treatment of this fatal disease is limited to chemotherapy using benzimidazoles and surgical intervention, with frequent disease recurrence in cases without radical surgery. Elucidating the molecular mechanisms underlying E. multilocularis infections and host-parasite interactions ultimately aids developing novel therapeutic options. This study explored an involvement of unfolded protein response (UPR) and endoplasmic reticulum-stress (ERS) during E. multilocularis infection in mice. METHODS E. multilocularis- and mock-infected C57BL/6 mice were subdivided into vehicle, albendazole (ABZ) and anti-programmed death ligand 1 (αPD-L1) treated groups. To mimic a chronic infection, treatments of mice started six weeks post i.p. infection and continued for another eight weeks. Liver tissue was then collected to examine inflammatory cytokines and the expression of UPR- and ERS-related genes. RESULTS E. multilocularis infection led to an upregulation of UPR- and ERS-related proteins in the liver, including ATF6, CHOP, GRP78, ERp72, H6PD and calreticulin, whilst PERK and its target eIF2α were not affected, and IRE1α and ATF4 were downregulated. ABZ treatment in E. multilocularis infected mice reversed, or at least tended to reverse, these protein expression changes to levels seen in mock-infected mice. Furthermore, ABZ treatment reversed the elevated levels of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α and interferon (IFN)-γ in the liver of infected mice. Similar to ABZ, αPD-L1 immune-treatment tended to reverse the increased CHOP and decreased ATF4 and IRE1α expression levels. CONCLUSIONS AND SIGNIFICANCE AE caused chronic inflammation, UPR activation and ERS in mice. The E. multilocularis-induced inflammation and consecutive ERS was ameliorated by ABZ and αPD-L1 treatment, indicating their effectiveness to inhibit parasite proliferation and downregulate its activity status. Neither ABZ nor αPD-L1 themselves affected UPR in control mice. Further research is needed to elucidate the link between inflammation, UPR and ERS, and if these pathways offer potential for improved therapies of patients with AE.
Collapse
Affiliation(s)
- Michael Weingartner
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Simon Stücheli
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Fadi Jebbawi
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Bruno Gottstein
- Institute for Infectious Diseases, Faculty of Medicine, University of Bern, Bern, Switzerland
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Guido Beldi
- Department of Visceral Surgery and Medicine, University Hospital of Bern, Bern, Switzerland
| | | | - Junhua Wang
- Institute for Infectious Diseases, Faculty of Medicine, University of Bern, Bern, Switzerland
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
18
|
Aslani MR, Amani M, Masrori N, Boskabady MH, Ebrahimi HA, Chodari L. Crocin attenuates inflammation of lung tissue in ovalbumin-sensitized mice by altering the expression of endoplasmic reticulum stress markers. Biofactors 2022; 48:204-215. [PMID: 34856021 DOI: 10.1002/biof.1809] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum (ER) stress plays a pivotal role in the pathogenesis of asthma. The present study aimed to investigate the reducing or suppressing effects of crocin in ovalbumin (OVA)-sensitized mice on ER stress markers. Mice were divided into six groups (n = 5 per group) including control, OVA-sensitized (OVA), OVA-treated crocin (OVA-Cr25, OVA-Cr50, and OVA-Cr100 mg/kg), and OVA-treated dexamethasone (1 mg/kg), (OVA-Dexa) groups. Animals 5 later groups were sensitized to OVA and the treatment groups received intraperitoneally crocin/dexamethasone in the last 5 days of the model. At the end of the study, lung tissue was evaluated for airway inflammation, caspase 12 and CHOP protein levels, and expression of ER stress markers using real-time-PCR. Sensitization with OVA significantly caused airway inflammation and induction of ER stress in mice compared to the control group based on the elevated inflammatory cells and ER stress markers in the lung tissue. Treatment with crocin and dexamethasone reduced airway inflammation and suppressed ER stress markers. Interestingly, in the OVA-Cr100 group, the suppressive effects on ER stress apoptotic markers were comparable to the OVA-Dexa group. The results suggest that crocin mediates maladaptive ER stress conditions possibly by creating adaptive ER stress status and driving protein folding correctly.
Collapse
Affiliation(s)
- Mohammad Reza Aslani
- Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mojtaba Amani
- Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Neghin Masrori
- Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Hossein Boskabady
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Ali Ebrahimi
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Leila Chodari
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
19
|
Pathinayake PS, Waters DW, Nichol KS, Brown AC, Reid AT, Hsu ACY, Horvat JC, Wood LG, Baines KJ, Simpson JL, Gibson PG, Hansbro PM, Wark PAB. Endoplasmic reticulum-unfolded protein response signalling is altered in severe eosinophilic and neutrophilic asthma. Thorax 2021; 77:443-451. [PMID: 34510013 DOI: 10.1136/thoraxjnl-2020-215979] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 07/06/2021] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The significance of endoplasmic reticulum (ER) stress in asthma is unclear. Here, we demonstrate that ER stress and the unfolded protein response (UPR) are related to disease severity and inflammatory phenotype. METHODS Induced sputum (n=47), bronchial lavage (n=23) and endobronchial biopsies (n=40) were collected from participants with asthma with varying disease severity, inflammatory phenotypes and from healthy controls. Markers for ER stress and UPR were assessed. These markers were also assessed in established eosinophilic and neutrophilic murine models of asthma. RESULTS Our results demonstrate increased ER stress and UPR pathways in asthma and these are related to clinical severity and inflammatory phenotypes. Genes associated with ER protein chaperone (BiP, CANX, CALR), ER-associated protein degradation (EDEM1, DERL1) and ER stress-induced apoptosis (DDIT3, PPP1R15A) were dysregulated in participants with asthma and are associated with impaired lung function (forced expiratory volume in 1 s) and active eosinophilic and neutrophilic inflammation. ER stress genes also displayed a significant correlation with classic Th2 (interleukin-4, IL-4/13) genes, Th17 (IL-17F/CXCL1) genes, proinflammatory (IL-1b, tumour necrosis factor α, IL-8) genes and inflammasome activation (NLRP3) in sputum from asthmatic participants. Mice with allergic airway disease (AAD) and severe steroid insensitive AAD also showed increased ER stress signalling in their lungs. CONCLUSION Heightened ER stress is associated with severe eosinophilic and neutrophilic inflammation in asthma and may play a crucial role in the pathogenesis of asthma.
Collapse
Affiliation(s)
- Prabuddha S Pathinayake
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia
| | - David W Waters
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Kristy S Nichol
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Alexandra C Brown
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Andrew T Reid
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Alan Chen-Yu Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Peter G Gibson
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia.,NHMRC Centre for Clinical Research Excellence in Severe Asthma, New Lambton Heights, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia.,Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia .,Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia.,NHMRC Centre for Clinical Research Excellence in Severe Asthma, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
20
|
Sweeter JM, Kudrna K, Hunt K, Thomes P, Dickey BF, Brody SL, Dickinson JD. Autophagy of mucin granules contributes to resolution of airway mucous metaplasia. Sci Rep 2021; 11:13037. [PMID: 34158522 PMCID: PMC8219712 DOI: 10.1038/s41598-021-91932-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 06/01/2021] [Indexed: 12/21/2022] Open
Abstract
Exacerbations of muco-obstructive airway diseases such as COPD and asthma are associated with epithelial changes termed mucous metaplasia (MM). Many molecular pathways triggering MM have been identified; however, the factors that regulate resolution are less well understood. We hypothesized that the autophagy pathway is required for resolution of MM by eliminating excess non-secreted intracellular mucin granules. We found increased intracellular levels of mucins Muc5ac and Muc5b in mice deficient in autophagy regulatory protein, Atg16L1, and that this difference was not due to defects in the known baseline or stimulated mucin secretion pathways. Instead, we found that, in mucous secretory cells, Lc3/Lamp1 vesicles colocalized with mucin granules particularly adjacent to the nucleus, suggesting that some granules were being eliminated in the autophagy pathway rather than secreted. Using a mouse model of MM resolution, we found increased lysosomal proteolytic activity that peaked in the days after mucin production began to decline. In purified lysosomal fractions, Atg16L1-deficient mice had reduced proteolytic degradation of Lc3 and Sqstm1 and persistent accumulation of mucin granules associated with impaired resolution of mucous metaplasia. In normal and COPD derived human airway epithelial cells (AECs), activation of autophagy by mTOR inhibition led to a reduction of intracellular mucin granules in AECs. Our findings indicate that during peak and resolution phases of MM, autophagy activity rather than secretion is required for elimination of some remaining mucin granules. Manipulation of autophagy activation offers a therapeutic target to speed resolution of MM in airway disease exacerbations.
Collapse
Affiliation(s)
- J M Sweeter
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - K Kudrna
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - K Hunt
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - P Thomes
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - B F Dickey
- Department of Pulmonary Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - S L Brody
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - J D Dickinson
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
21
|
Wang S, Jiang Z, Li L, Zhang J, Zhang C, Shao C. Ameliorative effects of eosinophil deficiency on immune response, endoplasmic reticulum stress, apoptosis, and autophagy in fungus-induced allergic lung inflammation. Respir Res 2021; 22:173. [PMID: 34098934 PMCID: PMC8186139 DOI: 10.1186/s12931-021-01770-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 06/04/2021] [Indexed: 01/21/2023] Open
Abstract
Background Respiratory fungal exposure is known to be associated with various allergic pulmonary disorders. Eosinophils have been implicated in tissue homeostasis of allergic inflammation as both destructive effector cells and immune regulators. What contributions eosinophils have in Aspergillus fumigatus (Af)-induced allergic lung inflammation is worthy of investigating. Methods We established the Af-exposed animal asthmatic model using eosinophil-deficient mice, ∆dblGATA1 mice. Airway inflammation was assessed by histopathological examination and total cell count of bronchoalveolar lavage fluid (BALF). The protein level in BALF and lung mRNA level of type 2 cytokines IL-4, IL-5, and IL-13 were detected by ELISA and qRT-PCR. We further studied the involvement of endoplasmic reticulum (ER) stress, apoptosis, and autophagy by western blots, qRT-PCR, immunofluorescence, TUNEL, or immunohistochemistry. RNA-Seq analysis was utilized to analyze the whole transcriptome of Af-exposed ∆dblGATA1 mice. Results Hematoxylin and eosin (HE) staining and periodic acid–Schiff staining (PAS) showed that airway inflammation and mucus production were alleviated in Af-challenged ∆dblGATA1 mice compared with wild-type controls. The protein and mRNA expressions of IL-4, IL-5, and IL-13 were reduced in the BALF and lung tissues in Af-exposed ∆dblGATA1 mice. The results demonstrated that the significantly increased ER stress markers (GRP78 and CHOP) and apoptosis executioner caspase proteases (cleaved caspase-3 and cleaved caspase-7) in Af-exposed wild-type mice were all downregulated remarkably in the lungs of ∆dblGATA1 mice with Af challenge. In addition, the lung autophagy in Af-exposed ∆dblGATA1 mice was found elevated partially, manifesting as higher expression of LC3-II/LC3-I and beclin1, lower p62, and downregulated Akt/mTOR pathway compared with Af-exposed wild-type mice. Additionally, lung RNA-seq analysis of Af-exposed ∆dblGATA1 mice showed that biological processes about chemotaxis of lymphocytes, neutrophils, or eosinophils were enriched but without statistical significance. Conclusions In summary, eosinophils play an essential role in the pathogenesis of Af-exposed allergic lung inflammation, whose deficiency may have relation to the attenuation of type 2 immune response, alleviation of ER stress and apoptosis, and increase of autophagy. These findings suggest that anti-eosinophils therapy may provide a promising direction for fungal-induced allergic pulmonary diseases.
Collapse
Affiliation(s)
- Sijiao Wang
- 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
| | - Liyang Li
- 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
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, 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
|
22
|
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
|
23
|
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: 7.3] [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
|
24
|
A new horizon for the old antibacterial drug clofoctol. Drug Discov Today 2021; 26:1302-1310. [PMID: 33581321 DOI: 10.1016/j.drudis.2021.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/16/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023]
Abstract
The synthetic antibacterial drug clofoctol (CFT) has long been used to treat respiratory tract infections in Europe. In recent years, the drug was found to target two biologically important proteins, the Cdc7/Dbf4 protein kinase complex and the mRNA-binding protein cold shock domain containing E1 (CSDE1), also known as upstream-of-N-Ras protein (UNR). These interactions are at the origin of the antitumor activity of CFT, recently evidenced in prostate cancer and neuroglioma. Drug-protein binding models provide a structural basis to guide the design of more potent anticancer compounds. A renewed interest in CFT can be anticipated for the treatment of cancers, and possibly Coronavirus 2019 (COVID-19).
Collapse
|
25
|
Chen X, Cubillos-Ruiz JR. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer 2021; 21:71-88. [PMID: 33214692 PMCID: PMC7927882 DOI: 10.1038/s41568-020-00312-2] [Citation(s) in RCA: 737] [Impact Index Per Article: 184.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Protein handling, modification and folding in the endoplasmic reticulum (ER) are tightly regulated processes that determine cell function, fate and survival. In several tumour types, diverse oncogenic, transcriptional and metabolic abnormalities cooperate to generate hostile microenvironments that disrupt ER homeostasis in malignant and stromal cells, as well as infiltrating leukocytes. These changes provoke a state of persistent ER stress that has been demonstrated to govern multiple pro-tumoural attributes in the cancer cell while dynamically reprogramming the function of innate and adaptive immune cells. Aberrant activation of ER stress sensors and their downstream signalling pathways have therefore emerged as key regulators of tumour growth and metastasis as well as response to chemotherapy, targeted therapies and immunotherapy. In this Review, we discuss the physiological inducers of ER stress in the tumour milieu, the interplay between oncogenic signalling and ER stress response pathways in the cancer cell and the profound immunomodulatory effects of sustained ER stress responses in tumours.
Collapse
Affiliation(s)
- Xi Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| | - Juan R Cubillos-Ruiz
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
26
|
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 PMCID: PMC12042654 DOI: 10.1165/rcmb.2019-0235tr] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [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
|
27
|
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: 2.6] [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
|
28
|
Dolly A, Dumas J, Servais S. Cancer cachexia and skeletal muscle atrophy in clinical studies: what do we really know? J Cachexia Sarcopenia Muscle 2020; 11:1413-1428. [PMID: 33053604 PMCID: PMC7749617 DOI: 10.1002/jcsm.12633] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/24/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Research investigators have shown a growing interest in investigating alterations underlying skeletal muscle wasting in patients with cancer. However, skeletal muscle dysfunctions associated with cancer cachexia have mainly been studied in preclinical models. In the present review, we summarize the results of clinical studies in which skeletal muscle biopsies were collected from cachectic vs. non-cachectic cancer patients. Most of these studies suggest the presence of significant physiological alterations in skeletal muscle from cachectic cancer patients. We suggest a hypothesis, which connects structural and metabolic parameters that may, at least in part, be responsible for the skeletal muscle atrophy characteristic of cancer cachexia. Finally, we discuss the importance of a better standardization of the diagnostic criteria for cancer cachexia, as well as the requirement for additional clinical studies to improve the robustness of these conclusions.
Collapse
Affiliation(s)
- Adeline Dolly
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| | - Jean‐François Dumas
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| | - Stéphane Servais
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| |
Collapse
|
29
|
Luthers CR, Dunn TM, Snow AL. ORMDL3 and Asthma: Linking Sphingolipid Regulation to Altered T Cell Function. Front Immunol 2020; 11:597945. [PMID: 33424845 PMCID: PMC7793773 DOI: 10.3389/fimmu.2020.597945] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Orosomucoid like 3 (ORMDL3) encodes an ER-resident transmembrane protein that regulates the activity of serine palmitoyltransferase (SPT), the first and rate-limiting enzyme for sphingolipid biosynthesis in cells. A decade ago, several genome wide association studies revealed single nucleotide polymorphisms associated with increased ORMDL3 protein expression and susceptibility to allergic asthma. Since that time, numerous studies have investigated how altered ORMDL3 expression might predispose to asthma and other autoimmune/inflammatory diseases. In this brief review, we focus on growing evidence suggesting that heightened ORMDL3 expression specifically in CD4+ T lymphocytes, the central orchestrators of adaptive immunity, constitutes a major underlying mechanism of asthma pathogenesis by skewing their differentiation and function. Furthermore, we explore how sphingolipid modulation in T cells might be responsible for these effects, and how further studies may interrogate this intriguing hypothesis.
Collapse
Affiliation(s)
- Christopher R Luthers
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Teresa M Dunn
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
30
|
Martinez-Carrasco R, Argüeso P, Fini ME. Dynasore protects ocular surface mucosal epithelia subjected to oxidative stress by maintaining UPR and calcium homeostasis. Free Radic Biol Med 2020; 160:57-66. [PMID: 32791188 PMCID: PMC7704702 DOI: 10.1016/j.freeradbiomed.2020.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/02/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
The mucosal epithelia of the ocular surface protect against external threats to the eye. Using a model of human stratified corneal epithelial cells with mucosal differentiation, we previously demonstrated that a small molecule inhibitor of dynamin GTPases, dynasore, prevents damage to cells and their transcellular barriers when subjected to oxidative stress. Investigating mechanisms, we now report the novel finding that dynasore acts by maintaining Ca+2 homeostasis, thereby inhibiting the PERK branch of the unfolded protein response (UPR) that promotes cell death. Dynasore was found to protect mitochondria by preventing mitochondrial permeability transition pore opening (mPTP), but, unlike reports using other systems, this was not mediated by dynamin family member DRP1. Necrostatin-1, an inhibitor of RIPK1 and lytic forms of programmed cell death, also inhibited mPTP opening and further protected the plasma membrane barrier. Significantly, necrostatin-1 did not protect the mucosal barrier. Oxidative stress increased mRNA for sXBP1, a marker of the IRE1 branch of the UPR, and CHOP, a marker of the PERK branch. It also stimulated phosphorylation of eIF2α, the upstream regulator of CHOP, as well as an increase in intracellular Ca2+. Dynasore selectively inhibited the increase in PERK branch markers, and also prevented the increase intracellular Ca2+ in response to oxidative stress. The increase in PERK branch markers were also inhibited when cells were treated with the cell permeable Ca2+ chelator, BAPTA-AM. To our knowledge, this is the first time that dynasore has been shown to have an effect on the UPR and suggests therapeutic applications.
Collapse
Affiliation(s)
- Rafael Martinez-Carrasco
- New England Eye Center of Tufts Medical Center, Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - Pablo Argüeso
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - M Elizabeth Fini
- New England Eye Center of Tufts Medical Center, Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA; Program in Pharmacology and Drug Development, Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|
31
|
Abstract
Proteins succumb to numerous post-translational modifications (PTMs). These relate to enzymatic or non-enzymatic reactions taking place in either the intracellular or extracellular compartment. While intracellular oxidative changes are mainly due to redox stress, extracellular PTMs may be induced in an inflammatory micro milieu that is rich in reactive species. The increasing recognition of oxidative modifications as a causing agent or side-effect of pathophysiological states and diseases puts oxidative PTMS (oxPTMs) into the spotlight of inflammation research. Pathological hyper-modification of proteins can lead to accumulation, aggregation, cell stress, altered antigenic peptides, and damage-associated molecular pattern (DAMP)-like recognition by host immunity. Such processes are linked to cardiovascular disease and autoinflammation. At the same time, a detailed understanding of the mechanisms governing inflammatory responses to oxPTMs may capitalize on new therapeutic routes for enhancing adaptive immune responses as needed, for instance, in oncology. We here summarize some of the latest developments of oxPTMs in disease diagnosis and therapy. Potential target proteins and upcoming technologies, such as gas plasmas, are outlined for future research that may aid in identifying the molecular basis of immunogenic vs. tolerogenic oxPTMs.
Collapse
|
32
|
Gomez JL, Chen A, Diaz MP, Zirn N, Gupta A, Britto C, Sauler M, Yan X, Stewart E, Santerian K, Grant N, Liu Q, Fry R, Rager J, Cohn L, Alexis N, Chupp GL. A Network of Sputum MicroRNAs Is Associated with Neutrophilic Airway Inflammation in Asthma. Am J Respir Crit Care Med 2020; 202:51-64. [PMID: 32255668 PMCID: PMC7328332 DOI: 10.1164/rccm.201912-2360oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/06/2020] [Indexed: 01/06/2023] Open
Abstract
Rationale: MicroRNAs are potent regulators of biologic systems that are critical to tissue homeostasis. Individual microRNAs have been identified in airway samples. However, a systems analysis of the microRNA-mRNA networks present in the sputum that contribute to airway inflammation in asthma has not been published.Objectives: Identify microRNA and mRNA networks in the sputum of patients with asthma.Methods: We conducted a genome-wide analysis of microRNA and mRNA in the sputum from patients with asthma and correlated expression with clinical phenotypes. Weighted gene correlation network analysis was implemented to identify microRNA networks (modules) that significantly correlate with clinical features of asthma and mRNA expression networks. MicroRNA expression in peripheral blood neutrophils and lymphocytes and in situ hybridization of the sputum were used to identify the cellular sources of microRNAs. MicroRNA expression obtained before and after ozone exposure was also used to identify changes associated with neutrophil counts in the airway.Measurements and Main Results: Six microRNA modules were associated with clinical features of asthma. A single module (nely) was associated with a history of hospitalizations, lung function impairment, and numbers of neutrophils and lymphocytes in the sputum. Of the 12 microRNAs in the nely module, hsa-miR-223-3p was the highest expressed microRNA in neutrophils and was associated with increased neutrophil counts in the sputum in response to ozone exposure. Multiple microRNAs in the nely module correlated with two mRNA modules enriched for TLR (Toll-like receptor) and T-helper cell type 17 (Th17) signaling and endoplasmic reticulum stress. hsa-miR-223-3p was a key regulator of the TLR and Th17 pathways in the sputum of subjects with asthma.Conclusions: This study of sputum microRNA and mRNA expression from patients with asthma demonstrates the existence of microRNA networks and genes that are associated with features of asthma severity. Among these, hsa-miR-223-3p, a neutrophil-derived microRNA, regulates TLR/Th17 signaling and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Jose L. Gomez
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Ailu Chen
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Maria Paula Diaz
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Nicholas Zirn
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Amolika Gupta
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Clemente Britto
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Xiting Yan
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Emma Stewart
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Kyle Santerian
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Nicole Grant
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Qing Liu
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Rebecca Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina; and
| | - Julia Rager
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina; and
| | - Lauren Cohn
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Neil Alexis
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina
| | - Geoffrey L. Chupp
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
33
|
Aslani MR, Ghobadi H, Panahpour H, Ahmadi M, Khaksar M, Heidarzadeh M. Modification of lung endoplasmic reticulum genes expression and NF-kB protein levels in obese ovalbumin-sensitized male and female rats. Life Sci 2020; 247:117446. [PMID: 32081662 DOI: 10.1016/j.lfs.2020.117446] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 12/21/2022]
Abstract
AIMS Previous studies showed a close relationship between obesity and asthma. In this study, we investigated the expression of endoplasmic reticulum (ER) stress genes in the lung tissue of obese ovalbumin (OVA)-sensitized male and female rats. MAIN METHODS The rats were divided into eight groups (n = 5 per group) as follows: female and male rats fed with normal diet (FND and MND, respectively), female and male OVA-sensitized rats fed with normal diet (F-OND and M-OND, respectively), female and male rats fed with high-fat diet (F-HFD and M-HFD, respectively), female and male OVA-sensitized rats fed with high-fat diet (F-OHFD and M-OHFD, respectively). All rats were fed with a high-fat diet or standard pelts for 8 weeks, and for another 4 weeks, they were sensitized by OVA or saline. At the end of the study, lung tissue NF-kB protein level was assessed, and ER stress markers genes expression was determined by Real Time-PCR. KEY FINDING OVA-sensitization and diet-induced obesity caused the curve of methacholine concentration-response to shift to the left. In addition, the results indicated that the EC50 (the effective concentration of methacholine generating 50% of peak response) in F-OHFD rats was statistically lower than that of the M-OHFD group (p < 0.05). Moreover, the results showed that diet-induced obesity increased the expression of ATF4, ATF6, GRP78, XBP-1, and CHOP as well as the protein level of NF-kB in this experimental model of asthma, markedly in the F-OHFD group. SIGNIFICANCE The results suggest that ER stress may be involved in the pathogenesis of asthma observed in obese OVA-sensitized rats, especially in the female animals.
Collapse
Affiliation(s)
- Mohammad Reza Aslani
- Lung Inflammatory Diseases Research Center, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran; Neurogenetic Inflammation Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hassan Ghobadi
- Department of Internal Medicine (Pulmonary Division), School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Hamdollah Panahpour
- Physiology Department, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mahdi Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Khaksar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Heidarzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
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: 7.6] [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
|
35
|
Bartoszewski R, Gebert M, Janaszak-Jasiecka A, Cabaj A, Króliczewski J, Bartoszewska S, Sobolewska A, Crossman DK, Ochocka R, Kamysz W, Kalinowski L, Dąbrowski M, Collawn JF. Genome-wide mRNA profiling identifies RCAN1 and GADD45A as regulators of the transitional switch from survival to apoptosis during ER stress. FEBS J 2020; 287:2923-2947. [PMID: 31880863 DOI: 10.1111/febs.15195] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/26/2019] [Accepted: 12/23/2019] [Indexed: 12/17/2022]
Abstract
Endoplasmic reticulum (ER) stress conditions promote a cellular adaptive mechanism called the unfolded protein response (UPR) that utilizes three stress sensors, inositol-requiring protein 1, protein kinase RNA-like ER kinase, and activating transcription factor 6. These sensors activate a number of pathways to reduce the stress and facilitate cell survival. While much is known about the mechanisms involved that modulate apoptosis during chronic stress, less is known about the transition between the prosurvival and proapoptotic factors that determine cell fate. Here, we employed a genetic screen that utilized three different pharmacological stressors to induce ER stress in a human-immortalized airway epithelial cell line, immortalized human bronchial epithelial cells. We followed the stress responses over an 18-h time course and utilized real-time monitoring of cell survival, next-generation sequencing, and quantitative real-time PCR to identify and validate genes that were upregulated with all three commonly employed ER stressors, inhibitor of calpain 1, tunicamycin, and thapsigargin. growth arrest and DNA damage-inducible alpha (GADD45A), a proapoptotic factor, and regulator of calcineurin 1 (RCAN1) mRNAs were identified and verified by showing that small interfering RNA (siRNA) knockdown of GADD45A decreased CCAAT-enhancer-binding protein homologous protein (a.k.a DDIT3), BCL2-binding component 3 (a.k.a. BBC3), and phorbol-12-myristate-13-acetate-induced protein 1 expression, 3 proapoptotic factors, and increased cell viability during ER stress conditions, whereas siRNA knockdown of RCAN1 dramatically decreased cell viability. These results suggest that the relative levels of these two genes regulate cell fate decisions during ER stress independent of the type of ER stressor.
Collapse
Affiliation(s)
- Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Poland
| | - Magdalena Gebert
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Poland
| | | | - Aleksandra Cabaj
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Jarosław Króliczewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Poland
| | | | - Aleksandra Sobolewska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Poland
| | - David K Crossman
- Department of Genetics, Heflin Center for Genomic Science, University of Alabama at Birmingham, AL, USA
| | - Renata Ochocka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Poland
| | - Wojciech Kamysz
- Department of Inorganic Chemistry, Medical University of Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Poland
| | - Michał Dąbrowski
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, AL, USA
| |
Collapse
|
36
|
Cooper DM, Loxham M. Particulate matter and the airway epithelium: the special case of the underground? Eur Respir Rev 2019; 28:28/153/190066. [PMID: 31554704 PMCID: PMC9488653 DOI: 10.1183/16000617.0066-2019] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/23/2019] [Indexed: 11/25/2022] Open
Abstract
Airborne particulate matter (PM) is a leading driver of premature mortality and cardiopulmonary morbidity, associated with exacerbations of asthma and chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and lung cancer. The airway epithelium, as the principal site of PM deposition, is critical to the effects of, and initial response to, PM. A key mechanism by which PM exerts its effects is the generation of reactive oxygen species (ROS), inducing antioxidant and inflammatory responses in exposed epithelial cells. However, much of what is known about the effects of PM is based on research using particulates from urban air. PM from underground railways is compositionally highly distinct from urban PM, being rich in metals associated with wheel, rail and brake wear and electrical arcing and component wear, which endows underground PM with potent ROS-generating capacity. In addition, underground PM appears to be more inflammogenic than urban PM in epithelial cells, but there is a lack of research into effects on exposed individuals, especially those with underlying health conditions. This review summarises current knowledge about the effects of PM on the airway epithelium, how the effects of underground PM may be different to urban PM and the potential health consequences and mitigation strategies for commuters and workers in underground railways. Airborne particulate matter in underground railways is much more concentrated and metal-rich than that found above ground. The evidence surrounding what this might mean for effects on the airways of exposed commuters and staff is limited and inconsistent.http://bit.ly/2KtcorT
Collapse
Affiliation(s)
- Dawn M Cooper
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Matthew Loxham
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK .,NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK.,Southampton Marine and Maritime Institute, University of Southampton, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
37
|
Abstract
Current management of severe asthma relying either on guidelines (bulk approach) or on disease phenotypes (stratified approach) did not improve the burden of the disease. Several severe phenotypes are described: clinical, functional, morphological, inflammatory, molecular and microbiome-related. However, phenotypes do not necessarily relate to or give insights into the underlying pathogenetic mechanisms which are described by the disease endotypes. Based on the major immune-inflammatory pathway involved type-2 high, type-2 low and mixed endotypes are described for severe asthma, with several shared pathogenetic pathways such as genetic and epigenetic, metabolic, neurogenic and remodelling subtypes. The concept of multidimensional endotyping as un unbiased approach to severe asthma is discussed, together with new tools and targets facilitating the shift from the stratified to the precision medicine approach.
Collapse
|
38
|
A Novel Insight on Endotyping Heterogeneous Severe Asthma Based on Endoplasmic Reticulum Stress: Beyond the "Type 2/Non-Type 2 Dichotomy". Int J Mol Sci 2019; 20:ijms20030713. [PMID: 30736433 PMCID: PMC6386842 DOI: 10.3390/ijms20030713] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 01/28/2019] [Accepted: 02/02/2019] [Indexed: 12/21/2022] Open
Abstract
Severe asthma is an extremely heterogeneous clinical syndrome in which diverse cellular and molecular pathobiologic mechanisms exist, namely endotypes. The current system for endotyping severe asthma is largely based on inflammatory cellular profiles and related pathways, namely the dichotomy of type 2 response (resulting in eosinophilic inflammation) and non-type 2 response (reinforcing non-eosinophilic inflammation involving neutrophils or less inflammatory cells), forming the basis of a development strategy for novel therapies. Although specific subgroups of type 2 severe asthma patients may derive benefit from modern precision medicine targeting type 2 cytokines, there is no approved and effective therapeutic agent for non-type 2 severe asthma, which comprises nearly 50% of all asthma patients. Importantly, the critical implication of endoplasmic reticulum (ER) stress and unfolded protein response—in close relation with several pivotal cellular immune/inflammatory platforms including mitochondria, NLRP3 inflammasome, and phosphoinositide 3-kinase-δ—in the generation of corticosteroid resistance is now being increasingly demonstrated in numerous experimental settings of severe asthma. Consistent with these findings, recent clinical data from a large European severe asthma cohort, in which molecular phenotyping as well as diverse clinical and physiological parameters from severe asthmatic patients were incorporated, suggest a brand new framework for endotyping severe asthma in relation to ER-associated mitochondria and inflammasome pathways. These findings highlight the view that ER stress-associated molecular pathways may serve as a unique endotype of severe asthma, and thus present a novel insight into the current knowledge and future development of treatment to overcome corticosteroid resistance in heterogeneous severe asthma.
Collapse
|
39
|
Kim BK, Lee HS, Sohn KH, Lee SY, Cho SH, Park HW. Different Biological Pathways Are Up-regulated in the Elderly With Asthma: Sputum Transcriptomic Analysis. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2019; 11:104-115. [PMID: 30479081 PMCID: PMC6267191 DOI: 10.4168/aair.2019.11.1.104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/11/2018] [Accepted: 09/14/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Elderly asthma (EA) is increasing, but the pathogenesis is unclear. This study aimed to identify EA-related biological pathways by analyzing genome-wide gene expression profiles in sputum cells. METHODS A total of 3,156 gene probes with significantly differential expressions between EA and healthy elderly controls were used for a hierarchical clustering of genes to identify gene clusters. Gene set enrichment analysis provided biological information, with replication from Gene Expression Omnibus expression profiles. RESULTS Fifty-five EA patients and 10 elderly control subjects were enrolled. Two distinct gene clusters were found. Cluster 1 (n = 35) showed a lower eosinophil proportion in sputum and less severe airway obstruction compared to cluster 2 (n = 20). The replication data set also identified 2 gene clusters (clusters 1' and 2'). Among 5 gene sets significantly enriched in cluster 1 and 3 gene sets significantly enriched in cluster 2, we confirmed that 2 were significantly enriched in the replication data set (OXIDATIVE_PHOSPHORYLATION gene set in cluster 1 and EPITHELIAL MESENCHYMAL TRANSITION gene set in cluster 2'). CONCLUSIONS The findings of 2 distinct gene clusters in EA and different biological pathways in each gene cluster suggest 2 different pathogenesis mechanisms underlying EA.
Collapse
Affiliation(s)
- Byung Keun Kim
- Department of Internal Medicine, Korea University Medical Center Anam Hospital, Seoul, Korea
| | - Hyun Seung Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Kyoung Hee Sohn
- Department of Internal Medicine, KyungHee University Medical center, Seoul, Korea
| | - Suh Young Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Sang Heon Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Heung Woo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea.
| |
Collapse
|