1
|
Wang M, Fu L, Wang H, Tian L. Hotspots and Trends in Allergic Rhinitis Nasal Mucosa Studies: A Bibliometric Analysis (2010-2024). J Asthma Allergy 2025; 18:417-435. [PMID: 40115252 PMCID: PMC11922781 DOI: 10.2147/jaa.s503477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/03/2025] [Indexed: 03/23/2025] Open
Abstract
Purpose This study aims to conduct a bibliometric and visual analysis of the research on the nasal mucosa in allergic rhinitis (AR) and to explore its emerging trends, hotspots, and future development. Methods We comprehensively searched the Web of Science Core Collection (WoSCC) for literature related to the nasal mucosa in AR published between 2010 and 2024. Bibliometric and visual analyses were performed using CiteSpace, VOSviewer, and the R language. Results A total of 1124 relevant articles were included in this study, and the analysis showed that the number of articles in this field has been increasing year by year. China dominated the article output, followed by South Korea and Japan. American Journal of Rhinology & Allergy (69 articles) topped the list of publications; keyword analysis showed that "immune response", "inflammatory response", "autophagy", "NLRP3 inflammasome", and "miRNAs" are hotspots in this field. Conclusion Over the past decade, research related to the nasal mucosa in AR have gained growing interest. This study is the first to use visualization software and data mining information to conduct a bibliometric analysis in this particular field, thereby providing fresh perspectives on the research terrain.
Collapse
Affiliation(s)
- Meiya Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Linyou Fu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Huan Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| | - Li Tian
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
2
|
Li S, Guo M, Long Y, Cai Y, Zhao Y, Huang S, Yang H, Fan Y, Chen X, Jin X. PIWI-interacting RNA MIABEPIR regulates cerebral endothelial cell function via DAPK2 pathway in offspring following maternal immune activation. Clin Transl Med 2025; 15:e70260. [PMID: 40000424 PMCID: PMC11859124 DOI: 10.1002/ctm2.70260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Maternal immune activation (MIA) is recognised as a risk factor in the neurodevelopmental disorders. However, the precise molecular pathways through which MIA disrupts neurovascular function remain largely unexplored. Here, we identify a novel MIA-associated brain endothelial piRNA (MIABEPIR) involved in regulating BMEC function and BBB integrity. RNA microarray analysis of foetal brain tissue from MIA-exposed mice revealed significant changes in piRNA expression, including a marked upregulation of MIABEPIR upregulated piRNAs. Immunofluorescence and FISH confirmed that MIABEPIR is localised in the microvascular endothelial cells of the brain. MIABEPIR overexpression enhances BMEC proliferation and angiogenesis but disrupts BBB integrity. In vivo, intracranial administration of lentiviral MIABEPIR in foetal mice resulted in marked BBB disruption. Mechanistically, we identified DAPK2 as a downstream target of MIABEPIR, leading to its downregulation. This suppression of DAPK2 inhibits autophagy in BMECs, suggesting that MIABEPIR modulates endothelial cell autophagy through the DAPK2 pathway. Our findings reveal a novel piRNA-mediated regulatory mechanism in neurovascular function during MIA and highlight MIABEPIR's role in MIA-induced neurodevelopmental abnormalities. Targeting the MIABEPIR-DAPK2 axis represents a potential therapeutic strategy for addressing neurovascular dysfunction in neurodevelopmental disorders associated with maternal immune stress.
Collapse
Affiliation(s)
| | - Miao Guo
- School of MedicineNankai UniversityTianjinChina
| | - Yao Long
- School of MedicineNankai UniversityTianjinChina
| | - Yuang Cai
- School of MedicineNankai UniversityTianjinChina
| | - Ying Zhao
- School of MedicineNankai UniversityTianjinChina
| | | | | | | | - Xu Chen
- School of MedicineNankai UniversityTianjinChina
- Tianjin Central Hospital of Gynecology ObstetricsTianjinChina
- Tianjin Key Laboratory of Human Development and Reproductive RegulationTianjinChina
| | - Xin Jin
- School of MedicineNankai UniversityTianjinChina
- Tianjin Central Hospital of Gynecology ObstetricsTianjinChina
- Tianjin Key Laboratory of Human Development and Reproductive RegulationTianjinChina
| |
Collapse
|
3
|
Miyano T, Sera T, Sakamoto N. Pharmacological activation of TRPML1 enhances autophagy regulating hypertonicity and TGF-β-induced EMT in proximal tubular epithelial cells. Biochem Biophys Res Commun 2025; 750:151432. [PMID: 39893888 DOI: 10.1016/j.bbrc.2025.151432] [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: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Proximal tubular epithelial cells (PTECs) are central to maintaining kidney homeostasis. Under pathological conditions, such as ischemia or inflammation, PTECs promote profibrotic signals, including transforming growth factor (TGF)-β, and undergo epithelial-mesenchymal transition (EMT). EMT is characterized by decreased epithelial markers (e.g., E-cadherin) and increased mesenchymal markers (e.g., α-smooth muscle actin [α-SMA]), which promote myofibroblast activation and fibrosis progression. We previously demonstrated that hyperosmotic stress, characterized by elevated extracellular solute concentrations, induces EMT in PTECs. However, we observed that hyperosmotic stress simultaneously activates autophagy, a cellular process that has antagonistic effects on EMT, primarily mediated by transient receptor potential mucolipin 1 (TRPML1). However, the interplay between hyperosmotic stress-induced EMT and autophagy remains unclear. This study examined whether enhancing autophagy via TRPML1 activation could modulate EMT under hyperosmotic stress. Using the TRPML1 agonist ML-SA1, we observed a significantly increased autophagic flux, indicated by elevated LC3-II levels, without cytotoxic effects. Under hyperosmotic conditions, ML-SA1 further amplified autophagic flux in PTECs compared to hyperosmotic stress alone. Notably, this enhanced autophagy suppressed EMT by maintaining E-cadherin expression and reducing α-SMA levels. Furthermore, the ML-SA1-mediated autophagy enhancement attenuated EMT and profibrotic factor production in TGF-β-treated cells, suggesting a broader protective role beyond hyperosmotic stress. These findings reveal a novel interaction between hyperosmotic stress-induced autophagy and EMT, emphasizing TRPML1 activation's therapeutic potential to mitigate PTEC injury and fibrosis progression.
Collapse
Affiliation(s)
- Takashi Miyano
- Department of Medical and Robotic Engineering Design, Tokyo University of Science, Tokyo, Japan; Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan.
| | - Toshihiro Sera
- Department of Medical and Robotic Engineering Design, Tokyo University of Science, Tokyo, Japan
| | - Naoya Sakamoto
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
4
|
Kumar D, Karvas RM, Jones BR, McColl ER, Diveley E, Sukanta J, Surendra S, Kelly JC, Theunissen TW, Mysorekar IU. SARS-CoV-2 ORF3a Protein Impairs Syncytiotrophoblast Maturation, Alters ZO-1 Localization, and Shifts Autophagic Pathways in Trophoblast Cells and 3D Organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614931. [PMID: 39386577 PMCID: PMC11463380 DOI: 10.1101/2024.09.25.614931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
SARS-CoV-2 infection poses a significant risk to placental physiology, but its impact on placental homeostasis is not well understood. We and others have previously shown that SARS-CoV-2 can colonize maternal and fetal placental cells, yet the specific mechanisms remain unclear. In this study, we investigate ORF3a, a key accessory protein of SARS-CoV-2 that exhibits continuous mutations. Our findings reveal that ORF3a is present in placental tissue from pregnant women infected with SARS-CoV-2 and disrupts autophagic flux in placental cell lines and 3D stem-cell-derived trophoblast organoids (SC-TOs), impairing syncytiotrophoblast differentiation and trophoblast invasion. This disruption leads to protein aggregation in cytotrophoblasts (CTB) and activates secretory autophagy, increasing CD63+ extracellular vesicle secretion, along with ORF3a itself. ORF3a also compromises CTB barrier integrity by disrupting tight junctions via interaction with ZO-1, mediated by its PDZ-binding motif, SVPL. Co-localization of ORF3a and ZO-1 in SARS-CoV-2-infected human placental tissue supports our in vitro findings. Deleting the PDZ binding motif in the ORF3a protein (ORF3a-noPBM mutant) restored proper ZO-1 localization at the cell junctions in an autophagy-independent manner. Lastly, we demonstrate that constitutive ORF3a expression induces SC-TOs to transition towards a secretory autophagy pathway likely via the PBM motif, as the ORF3a-NoPBM mutants showed a significant lack of CD63 expression. This study demonstrates the functional impact of ORF3a on placental autophagy and reveals a new mechanism for the activation of secretory autophagy, which may lead to increased extracellular vesicle secretion. These findings provide a foundation for exploring therapeutic approaches targeting ORF3a, specifically focusing on its PBM region to block its interactions with host cellular proteins and limiting placental impact.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rowan M. Karvas
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110
| | - Brittany R. Jones
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eliza R. McColl
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Emily Diveley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110
| | - Jash Sukanta
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Alpert School of Medicine, Providence, RI 02903)
| | - Sharma Surendra
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX 77555
| | - Jeannie C. Kelly
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110
| | - Thorold W. Theunissen
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110
| | - Indira U. Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center of Aging, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Cui L, Yang R, Huo D, Li L, Qu X, Wang J, Wang X, Liu H, Chen H, Wang X. Streptococcus pneumoniae extracellular vesicles aggravate alveolar epithelial barrier disruption via autophagic degradation of OCLN (occludin). Autophagy 2024; 20:1577-1596. [PMID: 38497494 PMCID: PMC11210924 DOI: 10.1080/15548627.2024.2330043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/25/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024] Open
Abstract
Streptococcus pneumoniae (S. pneumoniae) represents a major human bacterial pathogen leading to high morbidity and mortality in children and the elderly. Recent research emphasizes the role of extracellular vesicles (EVs) in bacterial pathogenicity. However, the contribution of S. pneumoniae EVs (pEVs) to host-microbe interactions has remained unclear. Here, we observed that S. pneumoniae infections in mice led to severe lung injuries and alveolar epithelial barrier (AEB) dysfunction. Infections of S. pneumoniae reduced the protein expression of tight junction protein OCLN (occludin) and activated macroautophagy/autophagy in lung tissues of mice and A549 cells. Mechanically, S. pneumoniae induced autophagosomal degradation of OCLN leading to AEB impairment in the A549 monolayer. S. pneumoniae released the pEVs that could be internalized by alveolar epithelial cells. Through proteomics, we profiled the cargo proteins inside pEVs and found that these pEVs contained many virulence factors, among which we identified a eukaryotic-like serine-threonine kinase protein StkP. The internalized StkP could induce the phosphorylation of BECN1 (beclin 1) at Ser93 and Ser96 sites, initiating autophagy and resulting in autophagy-dependent OCLN degradation and AEB dysfunction. Finally, the deletion of stkP in S. pneumoniae completely protected infected mice from death, significantly alleviated OCLN degradation in vivo, and largely abolished the AEB disruption caused by pEVs in vitro. Overall, our results suggested that pEVs played a crucial role in the spread of S. pneumoniae virulence factors. The cargo protein StkP in pEVs could communicate with host target proteins and even hijack the BECN1 autophagy initiation pathway, contributing to AEB disruption and bacterial pathogenicity.Abbreviations: AEB: alveolarepithelial barrier; AECs: alveolar epithelial cells; ATG16L1: autophagy related 16 like 1; ATP:adenosine 5'-triphosphate; BafA1: bafilomycin A1; BBB: blood-brain barrier; CFU: colony-forming unit; co-IP: co-immunoprecipitation; CQ:chloroquine; CTRL: control; DiO: 3,3'-dioctadecylox-acarbocyanineperchlorate; DOX: doxycycline; DTT: dithiothreitol; ECIS: electricalcell-substrate impedance sensing; eGFP: enhanced green fluorescentprotein; ermR: erythromycin-resistance expression cassette; Ery: erythromycin; eSTKs: eukaryotic-like serine-threoninekinases; EVs: extracellular vesicles; HA: hemagglutinin; H&E: hematoxylin and eosin; HsLC3B: human LC3B; hpi: hours post-infection; IP: immunoprecipitation; KD: knockdown; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LC/MS: liquid chromatography-mass spectrometry; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MVs: membranevesicles; NC:negative control; NETs:neutrophil extracellular traps; OD: optical density; OMVs: outer membrane vesicles; PBS: phosphate-buffered saline; pEVs: S.pneumoniaeextracellular vesicles; protK: proteinase K; Rapa: rapamycin; RNAi: RNA interference; S.aureus: Staphylococcusaureus; SNF:supernatant fluid; sgRNA: single guide RNA; S.pneumoniae: Streptococcuspneumoniae; S.suis: Streptococcussuis; TEER: trans-epithelium electrical resistance; moi: multiplicity ofinfection; TEM:transmission electron microscope; TJproteins: tight junction proteins; TJP1/ZO-1: tight junction protein1; TSA: tryptic soy agar; WB: western blot; WT: wild-type.
Collapse
Affiliation(s)
- Luqing Cui
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Ruicheng Yang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Dong Huo
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Liang Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xinyi Qu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Jundan Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xinyi Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hulin Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| |
Collapse
|
6
|
Jia L, Liu X, Liu X, Guan Q, Tian Y, Li J, Zhao P. Bufei Yishen formula protects the airway epithelial barrier and ameliorates COPD by enhancing autophagy through the Sirt1/AMPK/Foxo3 signaling pathway. Chin Med 2024; 19:32. [PMID: 38413976 PMCID: PMC10900682 DOI: 10.1186/s13020-024-00905-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/17/2024] [Indexed: 02/29/2024] Open
Abstract
OBJECT Bufei Yishen formula (BYF), a traditional Chinese medicine alleviates COPD symptoms and suppresses airway epithelial inflammation. In this study, we determined whether BYF protects the airway epithelial barrier from destruction in COPD rats. METHODS The protective effects of BYF on the airway epithelial barrier were examined in a rat COPD model. BEAS-2B epithelial cells were exposed to cigarette smoke extract (CSE) to determine the effect of BYF on epithelial barrier function. Transcriptomic and network analyses were conducted to identify the protective mechanisms. RESULTS Oral BYF reduced the severity of COPD in rats by suppressing the decline in lung function, pathological changes, inflammation, and protected airway epithelial barrier function by upregulating apical junction proteins, including occludin (OCLN), zonula occludens (ZO)-1, and E-cadherin (E-cad). BYF treatment reduced epithelial permeability, and increased TEER as well as the apical junction proteins, OCLN, ZO-1, and E-cad in BEAS-2B cells exposed to CSE. Furthermore, 58 compounds identified in BYF were used to predict 421 potential targets. In addition, the expression of 572 differentially expressed genes (DEGs) was identified in CSE-exposed BEAS-2B cells. A network analysis of the 421 targets and 572 DEGs revealed that BYF regulates multiple pathways, of which the Sirt1, AMPK, Foxo3, and autophagy pathways may be the most important with respect to protective mechanisms. Moreover, in vitro experiments confirmed that nobiletin, one of the active compounds in BYF, increased apical junction protein levels, including OCLN, ZO-1, and E-cad. It also increased LC3B and phosphorylated AMPK levels and decreased the phosphorylation of FoxO3a. CONCLUSIONS BYF protects the airway epithelial barrier in COPD by enhancing autophagy through regulation of the SIRT1/AMPK/FOXO3 signaling pathway.
Collapse
Affiliation(s)
- Lidan Jia
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, 450046, Henan Province, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, 450046, Henan Province, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Xinguang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, 450046, Henan Province, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Qingzhou Guan
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, 450046, Henan Province, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, 450046, Henan Province, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China.
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, 450046, Henan Province, China.
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China.
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, 450046, Henan Province, China.
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| |
Collapse
|
7
|
Ling C, Versloot CJ, Arvidsson Kvissberg ME, Hu G, Swain N, Horcas-Nieto JM, Miraglia E, Thind MK, Farooqui A, Gerding A, van Eunen K, Koster MH, Kloosterhuis NJ, Chi L, ChenMi Y, Langelaar-Makkinje M, Bourdon C, Swann J, Smit M, de Bruin A, Youssef SA, Feenstra M, van Dijk TH, Thedieck K, Jonker JW, Kim PK, Bakker BM, Bandsma RHJ. Rebalancing of mitochondrial homeostasis through an NAD +-SIRT1 pathway preserves intestinal barrier function in severe malnutrition. EBioMedicine 2023; 96:104809. [PMID: 37738832 PMCID: PMC10520344 DOI: 10.1016/j.ebiom.2023.104809] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND The intestine of children with severe malnutrition (SM) shows structural and functional changes that are linked to increased infection and mortality. SM dysregulates the tryptophan-kynurenine pathway, which may impact processes such as SIRT1- and mTORC1-mediated autophagy and mitochondrial homeostasis. Using a mouse and organoid model of SM, we studied the repercussions of these dysregulations on malnutrition enteropathy and the protective capacity of maintaining autophagy activity and mitochondrial health. METHODS SM was induced through feeding male weanling C57BL/6 mice a low protein diet (LPD) for 14-days. Mice were either treated with the NAD+-precursor, nicotinamide; an mTORC1-inhibitor, rapamycin; a SIRT1-activator, resveratrol; or SIRT1-inhibitor, EX-527. Malnutrition enteropathy was induced in enteric organoids through amino-acid deprivation. Features of and pathways to malnutrition enteropathy were examined, including paracellular permeability, nutrient absorption, and autophagic, mitochondrial, and reactive-oxygen-species (ROS) abnormalities. FINDINGS LPD-feeding and ensuing low-tryptophan availability led to villus atrophy, nutrient malabsorption, and intestinal barrier dysfunction. In LPD-fed mice, nicotinamide-supplementation was linked to SIRT1-mediated activation of mitophagy, which reduced damaged mitochondria, and improved intestinal barrier function. Inhibition of mTORC1 reduced intestinal barrier dysfunction and nutrient malabsorption. Findings were validated and extended using an organoid model, demonstrating that resolution of mitochondrial ROS resolved barrier dysfunction. INTERPRETATION Malnutrition enteropathy arises from a dysregulation of the SIRT1 and mTORC1 pathways, leading to disrupted autophagy, mitochondrial homeostasis, and ROS. Whether nicotinamide-supplementation in children with SM could ameliorate malnutrition enteropathy should be explored in clinical trials. FUNDING This work was supported by the Bill and Melinda Gates Foundation, the Sickkids Research Institute, the Canadian Institutes of Health Research, and the University Medical Center Groningen.
Collapse
Affiliation(s)
- Catriona Ling
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Christian J Versloot
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Matilda E Arvidsson Kvissberg
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Guanlan Hu
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nathan Swain
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - José M Horcas-Nieto
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Emily Miraglia
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mehakpreet K Thind
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Amber Farooqui
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Albert Gerding
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Karen van Eunen
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Mirjam H Koster
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Lijun Chi
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - YueYing ChenMi
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Miriam Langelaar-Makkinje
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Celine Bourdon
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jonathan Swann
- Faculty of Medicine, School of Human Development and Health, University of Southampton, United Kingdom; Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, United Kingdom
| | - Marieke Smit
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Alain de Bruin
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Centre, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Sameh A Youssef
- Department of Biomolecular Health Sciences, Dutch Molecular Pathology Centre, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Janssen Pharmaceutica Research and Development, 2340, Beerse, Belgium
| | - Marjon Feenstra
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Theo H van Dijk
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Kathrin Thedieck
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria; Freiburg Materials Research Center (FMF), University Freiburg, Freiburg, Germany
| | - Johan W Jonker
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Peter K Kim
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Barbara M Bakker
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands.
| | - Robert H J Bandsma
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, the Netherlands; Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
8
|
Nabavi-Rad A, Yadegar A, Sadeghi A, Aghdaei HA, Zali MR, Klionsky DJ, Yamaoka Y. The interaction between autophagy, Helicobacter pylori, and gut microbiota in gastric carcinogenesis. Trends Microbiol 2023; 31:1024-1043. [PMID: 37120362 PMCID: PMC10523907 DOI: 10.1016/j.tim.2023.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 05/01/2023]
Abstract
Chronic infection with Helicobacter pylori is the primary risk factor for the development of gastric cancer. Hindering our ability to comprehend the precise role of autophagy during H. pylori infection is the complexity of context-dependent autophagy signaling pathways. Recent and ongoing progress in understanding H. pylori virulence allows new frontiers of research for the crosstalk between autophagy and H. pylori. Novel approaches toward discovering autophagy signaling networks have further revealed their critical influence on the structure of gut microbiota and the metabolome. Here we intend to present a holistic view of the perplexing role of autophagy in H. pylori pathogenesis and carcinogenesis. We also discuss the intermediate role of autophagy in H. pylori-mediated modification of gut inflammatory responses and microbiota structure.
Collapse
Affiliation(s)
- Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Oita, Japan; Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX, USA; Research Center for Global and Local Infectious Diseases, Oita University, Oita, Japan.
| |
Collapse
|
9
|
Saha K, Subramenium Ganapathy A, Wang A, Michael Morris N, Suchanec E, Ding W, Yochum G, Koltun W, Nighot M, Ma T, Nighot P. Autophagy Reduces the Degradation and Promotes Membrane Localization of Occludin to Enhance the Intestinal Epithelial Tight Junction Barrier against Paracellular Macromolecule Flux. J Crohns Colitis 2023; 17:433-449. [PMID: 36219473 PMCID: PMC10069622 DOI: 10.1093/ecco-jcc/jjac148] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND AIMS Functional loss of the gut epithelium's paracellular tight junction [TJ] barrier and defective autophagy are factors potentiating inflammatory bowel disease [IBD]. Previously, we showed the role of autophagy in enhancing the intestinal TJ barrier via pore-forming claudin-2 degradation. How autophagy regulates the TJ barrier-forming proteins remains unknown. Here, we investigated the role of autophagy in the regulation of occludin, a principal TJ component involved in TJ barrier enhancement. RESULTS Autophagy induction using pharmacological activators and nutrient starvation increased total occludin levels in intestinal epithelial cells, mouse colonocytes and human colonoids. Autophagy induction enriched membrane occludin levels and reduced paracellular permeability of macromolecules. Autophagy-mediated TJ barrier enhancement was contingent on the presence of occludin as OCLN-/- nullified its TJ barrier-enhancing effect against macromolecular flux. Autophagy inhibited the constitutive degradation of occludin by preventing its caveolar endocytosis from the membrane and protected against inflammation-induced TJ barrier loss. Autophagy enhanced the phosphorylation of ERK-1/2 and inhibition of these kinases in Caco-2 cells and human colonic mucosa prevented the macromolecular barrier-enhancing effects of autophagy. In vivo, autophagy induction by rapamycin enhanced occludin levels in wild-type mouse intestines and protected against lipopolysaccharide- and tumour necrosis factor-α-induced TJ barrier loss. Disruption of autophagy with acute Atg7 knockout in adult mice decreased intestinal occludin levels, increasing baseline colonic TJ permeability and exacerbating the effect of experimental colitis. CONCLUSION Our data suggest a novel role of autophagy in promoting the intestinal TJ barrier by increasing occludin levels in an ERK1/2 mitogen-activated protein kinase-dependent mechanism.
Collapse
Affiliation(s)
- Kushal Saha
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Ashwinkumar Subramenium Ganapathy
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Alexandra Wang
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nathan Michael Morris
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Eric Suchanec
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Wei Ding
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Gregory Yochum
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Walter Koltun
- Division of Colon and Rectal Surgery, Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Meghali Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Thomas Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
10
|
Dietary Leucine Improves Fish Intestinal Barrier Function by Increasing Humoral Immunity, Antioxidant Capacity, and Tight Junction. Int J Mol Sci 2023; 24:ijms24054716. [PMID: 36902147 PMCID: PMC10003359 DOI: 10.3390/ijms24054716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
This study attempted to evaluate the possible impact and mechanism of leucine (Leu) on fish intestinal barrier function. One hundred and five hybrid Pelteobagrus vachelli ♀ × Leiocassis longirostris ♂ catfish were fed with six diets in graded levels of Leu 10.0 (control group), 15.0, 20.0, 25.0, 30.0, 35.0, and 40.0 g/kg diet for 56 days. Results showed that the intestinal activities of LZM, ACP, and AKP and contents of C3, C4, and IgM had positive linear and/or quadratic responses to dietary Leu levels. The mRNA expressions of itnl1, itnl2, c-LZM, g-LZM, and β-defensin increased linearly and/or quadratically (p < 0.05). The ROS, PC, and MDA contents had a negative linear and/or quadratic response, but GSH content and ASA, AHR, T-SOD, and GR activities had positive quadratic responses to dietary Leu levels (p < 0.05). No significant differences on the CAT and GPX activities were detected among treatments (p > 0.05). Increasing dietary Leu level linearly and/or quadratically increased the mRNA expressions of CuZnSOD, CAT, and GPX1α. The GST mRNA expression decreased linearly while the GCLC and Nrf2 mRNA expressions were not significantly affected by different dietary Leu levels. The Nrf2 protein level quadratically increased, whereas the Keap1 mRNA expression and protein level decreased quadratically (p < 0.05). The translational levels of ZO-1 and occludin increased linearly. No significant differences were indicated in Claudin-2 mRNA expression and protein level. The transcriptional levels of Beclin1, ULK1b, ATG5, ATG7, ATG9a, ATG4b, LC3b, and P62 and translational levels of ULK1, LC3Ⅱ/Ⅰ, and P62 linearly and quadratically decreased. The Beclin1 protein level was quadratically decreased with increasing dietary Leu levels. These results suggested that dietary Leu could improve fish intestinal barrier function by increasing humoral immunity, antioxidative capacities, and tight junction protein levels.
Collapse
|
11
|
Fu R, Jiang X, Li G, Zhu Y, Zhang H. Junctional complexes in epithelial cells: sentinels for extracellular insults and intracellular homeostasis. FEBS J 2022; 289:7314-7333. [PMID: 34453866 DOI: 10.1111/febs.16174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 01/13/2023]
Abstract
The cell-cell and cell-ECM junctions within the epithelial tissues are crucial anchoring structures that provide architectural stability, mechanical resistance, and permeability control. Their indispensable role as signaling hubs orchestrating cell shape-related changes such as proliferation, differentiation, migration, and apoptosis has also been well recognized. However, growing amount of evidence now suggests that the multitasking nature of epithelial junctions extends well beyond anchorage-dependent or cell shape change-related biological processes. In this review, we discuss the emerging roles of junctional complexes in regulating innate immune defense, stress resistance, and intracellular proteostasis of the epithelial cells, with emphasis on the upstream regulation of epithelial junctions on various aspects of the epithelial barrier.
Collapse
Affiliation(s)
- Rong Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| | - Xiaowan Jiang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| | - Gang Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| | - Yi Zhu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| | - Huimin Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, China
| |
Collapse
|
12
|
Alfonso-Pérez T, Baonza G, Herranz G, Martín-Belmonte F. Deciphering the interplay between autophagy and polarity in epithelial tubulogenesis. Semin Cell Dev Biol 2022; 131:160-172. [PMID: 35641407 DOI: 10.1016/j.semcdb.2022.05.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 12/14/2022]
Abstract
The Metazoan complexity arises from a primary building block, the epithelium, which comprises a layer of polarized cells that divide the organism into compartments. Most of these body compartments are organs formed by epithelial tubes that enclose an internal hollow space or lumen. Over the last decades, multiple studies have unmasked the paramount events required to form this lumen de novo. In epithelial cells, these events mainly involve recognizing external clues, establishing and maintaining apicobasal polarity, endo-lysosomal trafficking, and expanding the created lumen. Although canonical autophagy has been classically considered a catabolic process needed for cell survival, multiple studies have also emphasized its crucial role in epithelial polarity, morphogenesis and cellular homeostasis. Furthermore, non-canonical autophagy pathways have been recently discovered as atypical secretory routes. Both canonical and non-canonical pathways play essential roles in epithelial polarity and lumen formation. This review addresses how the molecular machinery for epithelial polarity and autophagy interplay in different processes and how autophagy functions influence lumenogenesis, emphasizing its role in the lumen formation key events.
Collapse
Affiliation(s)
- Tatiana Alfonso-Pérez
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo, Ochoa", CSIC-UAM, Madrid 28049, Spain; Ramon & Cajal Health Research Institute (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid 28034, Spain
| | - Gabriel Baonza
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo, Ochoa", CSIC-UAM, Madrid 28049, Spain
| | - Gonzalo Herranz
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo, Ochoa", CSIC-UAM, Madrid 28049, Spain; Ramon & Cajal Health Research Institute (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid 28034, Spain
| | - Fernando Martín-Belmonte
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo, Ochoa", CSIC-UAM, Madrid 28049, Spain; Ramon & Cajal Health Research Institute (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid 28034, Spain.
| |
Collapse
|
13
|
Glaesserella parasuis serotype 5 breaches the porcine respiratory epithelial barrier by inducing autophagy and blocking the cell membrane Claudin-1 replenishment. PLoS Pathog 2022; 18:e1010912. [PMID: 36228044 PMCID: PMC9595547 DOI: 10.1371/journal.ppat.1010912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/25/2022] [Accepted: 10/04/2022] [Indexed: 11/22/2022] Open
Abstract
Glaesserella parasuis (G. parasuis), the primary pathogen of Glässer's disease, colonizes the upper respiratory tract and can break through the epithelial barrier of the respiratory tract, leading to lung infection. However, the underlying mechanisms for this adverse effect remain unclear. The G. parasuis serotype 5 SQ strain (HPS5-SQ) infection decreased the integrity of piglets' lung Occludin and Claudin-1. Autophagy regulates the function of the epithelial barrier and tight junction proteins (TJs) expression. We tested the hypothesis that HPS5-SQ breaking through the porcine respiratory epithelial barrier was linked to autophagy and Claudin-1 degradation. When HPS5-SQ infected swine tracheal epithelial cells (STEC), autophagosomes encapsulated, and autolysosomes degraded oxidatively stressed mitochondria covered with Claudin-1. Furthermore, we found that autophagosomes encapsulating mitochondria resulted in cell membrane Claudin-1 being unable to be replenished after degradation and damaged the respiratory tract epithelial barrier. In conclusion, G. parasuis serotype 5 breaks through the porcine respiratory epithelial barrier by inducing autophagy and interrupting cell membrane Claudin-1 replenishment, clarifying the mechanism of the G. parasuis infection and providing a new potential target for drug design and vaccine development.
Collapse
|
14
|
Rovira M, Sereda R, Pladevall‐Morera D, Ramponi V, Marin I, Maus M, Madrigal‐Matute J, Díaz A, García F, Muñoz J, Cuervo AM, Serrano M. The lysosomal proteome of senescent cells contributes to the senescence secretome. Aging Cell 2022; 21:e13707. [PMID: 36087066 PMCID: PMC9577959 DOI: 10.1111/acel.13707] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/01/2022] [Accepted: 08/13/2022] [Indexed: 01/31/2023] Open
Abstract
Senescent cells accumulate in tissues over time, favoring the onset and progression of multiple age-related diseases. Senescent cells present a remarkable increase in lysosomal mass and elevated autophagic activity. Here, we report that two main autophagic pathways macroautophagy (MA) and chaperone-mediated autophagy (CMA) are constitutively upregulated in senescent cells. Proteomic analyses of the subpopulations of lysosomes preferentially engaged in each of these types of autophagy revealed profound quantitative and qualitative changes in senescent cells, affecting both lysosomal resident proteins and cargo proteins delivered to lysosomes for degradation. These studies have led us to identify resident lysosomal proteins that are highly augmented in senescent cells and can be used as novel markers of senescence, such as arylsulfatase ARSA. The abundant secretome of senescent cells, known as SASP, is considered their main pathological mediator; however, little is known about the mechanisms of SASP secretion. Some secretory cells, including melanocytes, use the small GTPase RAB27A to perform lysosomal secretion. We found that this process is exacerbated in the case of senescent melanoma cells, as revealed by the exposure of lysosomal membrane integral proteins LAMP1 and LAMP2 in their plasma membrane. Interestingly, a subset of SASP components, including cytokines CCL2, CCL3, CXCL12, cathepsin CTSD, or the protease inhibitor SERPINE1, are secreted in a RAB27A-dependent manner in senescent melanoma cells. Finally, proteins previously identified as plasma biomarkers of aging are highly enriched in the lysosomes of senescent cells, including CTSD. We conclude that the lysosomal proteome of senescent cells is profoundly reconfigured, and that some senescent cells can be highly active in lysosomal exocytosis.
Collapse
Affiliation(s)
- Miguel Rovira
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Rebecca Sereda
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineNew YorkNew YorkUSA
| | - David Pladevall‐Morera
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy AgingUniversity of CopenhagenCopenhagenDenmark
| | - Valentina Ramponi
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Ines Marin
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Mate Maus
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Julio Madrigal‐Matute
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Instituto Biomédico de Nutrición y SaludEldaSpain
| | - Antonio Díaz
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineNew YorkNew YorkUSA
| | - Fernando García
- Proteomics UnitSpanish National Cancer Research Center (CNIO)MadridSpain
| | - Javier Muñoz
- Proteomics UnitSpanish National Cancer Research Center (CNIO)MadridSpain
- Biocruces Bizkaia Health Research InstituteBarakaldoSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| | - Ana María Cuervo
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineNew YorkNew YorkUSA
| | - Manuel Serrano
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
| |
Collapse
|
15
|
Singh SB, Coffman CN, Varga MG, Carroll-Portillo A, Braun CA, Lin HC. Intestinal Alkaline Phosphatase Prevents Sulfate Reducing Bacteria-Induced Increased Tight Junction Permeability by Inhibiting Snail Pathway. Front Cell Infect Microbiol 2022; 12:882498. [PMID: 35694541 PMCID: PMC9177943 DOI: 10.3389/fcimb.2022.882498] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Tight junctions (TJs) are essential components of intestinal barrier integrity and protect the epithelium against passive paracellular flux and microbial translocation. Dysfunctional TJ leads to leaky gut, a condition associated with diseases including inflammatory bowel disease (IBD). Sulfate-Reducing Bacteria (SRB) are minor residents of the gut. An increased number of Desulfovibrio, the most predominant SRB, is observed in IBD and other diseases associated with leaky gut. However, it is not known whether Desulfovibrio contributes to leaky gut. We tested the hypothesis that Desulfovibrio vulgaris (DSV) may induce intestinal permeability in vitro. Snail, a transcription factor, disrupts barrier function by affecting TJ proteins such as occludin. Intestinal alkaline phosphatase (IAP), a host defense protein, protects epithelial barrier integrity. We tested whether DSV induced permeability in polarized Caco-2 cells via snail and if this effect was inhibited by IAP. Barrier integrity was assessed by measuring transepithelial electric resistance (TEER) and by 4kDa FITC-Dextran flux to determine paracellular permeability. We found that DSV reduced TEER, increased FITC-flux, upregulated snail protein expression, caused nuclear translocation of snail, and disrupted occludin staining at the junctions. DSV-induced permeability effects were inhibited in cells knocked down for snail. Pre-treatment of cells with IAP inhibited DSV-induced FITC flux and snail expression and DSV-mediated disruption of occludin staining. These data show that DSV, a resident commensal bacterium, can contribute to leaky gut and that snail may serve as a novel therapeutic target to mitigate DSV-induced effects. Taken together, our study suggests a novel underlying mechanism of association of Desulfovibrio bloom with diseases with increased intestinal permeability. Our study also underscores IAP as a novel therapeutic intervention for correcting SRB-induced leaky gut via inhibition of snail.
Collapse
Affiliation(s)
- Sudha B. Singh
- Biomedical Research Institute of New Mexico, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
| | - Cristina N. Coffman
- Biomedical Research Institute of New Mexico, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
| | - Matthew G. Varga
- Biomedical Research Institute of New Mexico, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
| | - Amanda Carroll-Portillo
- Division of Gastroenterology and Hepatology, Department of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Cody A. Braun
- Biomedical Research Institute of New Mexico, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
| | - Henry C. Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, University of New Mexico, Albuquerque, NM, United States
- Medicine Service, New Mexico Veterans Affairs (VA) Health Care System, Albuquerque, NM, United States
- *Correspondence: Henry C. Lin,
| |
Collapse
|
16
|
Qiu P, Liu L, Fang J, Zhang M, Wang H, Peng Y, Chen M, Liu J, Wang F, Zhao Q. Identification of Pharmacological Autophagy Regulators of Active Ulcerative Colitis. Front Pharmacol 2021; 12:769718. [PMID: 34925026 PMCID: PMC8672246 DOI: 10.3389/fphar.2021.769718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/15/2021] [Indexed: 11/29/2022] Open
Abstract
Background: Ulcerative colitis (UC) is a chronic recurrent disease of unknown etiology. Recently, it has been reported that autophagy-related gene polymorphism is closely associated with increased risk of UC, and the therapeutic effect of some UC drugs is mediated by regulating autophagy pathways. This study aims to identify pivotal autophagy-related regulators in UC pathogenesis and provide novel molecular targets for the treatment of active UC. Methods: Gene expression profiles and clinical information of active UC patients were obtained from GEO databases. CIBERSORT was adopted to evaluate the immune cell infiltration. We used weighted gene co-expression network analysis (WGCNA) and differential expression analysis to identify the pivotal modules and genes associated with active UC. Subsequently, we conducted validation in the validation set and explored its relationship with commonly used UC therapeutics. Results: 36 healthy controls and 46 active UC patients have been obtained from the training set of GSE53306, GSE87466, and GSE134025. There were 423 differentially expressed genes (DEGs) found, which dramatically enriched in autophagy-related pathways. And more infiltration of mast cells, activated T cells, dendritic cells, and M1 macrophages were observed in the intestinal mucosa of active UC, while more infiltration of resting immune cells and M2 macrophages in healthy controls. WGCNA indicated that the turquoise and blue modules were the critical modules. CASP1, SERPINA1, and CCL2 have been identified as the hub autophagy-related genes of active UC, after combining DEGs and 232 autophagy-related genes from HADb with the genes of turquoise and blue modules, respectively. We further verified that CASP1, SERPINA1, and CCL2 were positively associated with active UC and served as an autophagy-related biomarker for active UC. Moreover, increased SERPINA1 in the involved intestinal mucosa was reduced in patients with active UC who responded to golimumab or glucocorticoid therapy. But, neither CASP1, SERPINA1, and CCL2 were changed by treatment of 5-aminosalicylic acid (5-ASA) and azathioprine. Conclusion: CASP1, SERPINA1, and CCL2 are autophagy-related hub genes of active UC. And SERPINA1 may serve as a new pharmacological autophagy regulator of UC, which provides a new target for the use of small molecules targeting autophagy in the treatment of active UC.
Collapse
Affiliation(s)
- Peishan Qiu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Jun Fang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Meng Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Haizhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Yanan Peng
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Min Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Jing Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Fan Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan, China
| |
Collapse
|
17
|
Siva Sankar D, Dengjel J. Protein complexes and neighborhoods driving autophagy. Autophagy 2021; 17:2689-2705. [PMID: 33183148 PMCID: PMC8526019 DOI: 10.1080/15548627.2020.1847461] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/16/2020] [Accepted: 11/02/2020] [Indexed: 01/02/2023] Open
Abstract
Autophagy summarizes evolutionarily conserved, intracellular degradation processes targeting cytoplasmic material for lysosomal degradation. These encompass constitutive processes as well as stress responses, which are often found dysregulated in diseases. Autophagy pathways help in the clearance of damaged organelles, protein aggregates and macromolecules, mediating their recycling and maintaining cellular homeostasis. Protein-protein interaction networks contribute to autophagosome biogenesis, substrate loading, vesicular trafficking and fusion, protein translocations across membranes and degradation in lysosomes. Hypothesis-free proteomic approaches tremendously helped in the functional characterization of protein-protein interactions to uncover molecular mechanisms regulating autophagy. In this review, we elaborate on the importance of understanding protein-protein-interactions of varying affinities and on the strengths of mass spectrometry-based proteomic approaches to study these, generating new mechanistic insights into autophagy regulation. We discuss in detail affinity purification approaches and recent developments in proximity labeling coupled to mass spectrometry, which uncovered molecular principles of autophagy mechanisms.Abbreviations: AMPK: AMP-activated protein kinase; AP-MS: affinity purification-mass spectrometry; APEX2: ascorbate peroxidase-2; ATG: autophagy related; BioID: proximity-dependent biotin identification; ER: endoplasmic reticulum; GFP: green fluorescent protein; iTRAQ: isobaric tag for relative and absolute quantification; MS: mass spectrometry; PCA: protein-fragment complementation assay; PL-MS: proximity labeling-mass spectrometry; PtdIns3P: phosphatidylinositol-3-phosphate; PTM: posttranslational modification; PUP-IT: pupylation-based interaction tagging; RFP: red fluorescent protein; SILAC: stable isotope labeling by amino acids in cell culture; TAP: tandem affinity purification; TMT: tandem mass tag.
Collapse
Affiliation(s)
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
18
|
Regulation of Paneth Cell Function by RNA-Binding Proteins and Noncoding RNAs. Cells 2021; 10:cells10082107. [PMID: 34440876 PMCID: PMC8392049 DOI: 10.3390/cells10082107] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/20/2022] Open
Abstract
Paneth cells are specialized intestinal epithelial cells that are located at the base of small intestinal crypts and play a vital role in preserving the gut epithelium homeostasis. Paneth cells act as a safeguard from bacterial translocation across the epithelium and constitute the niche for intestinal stem cells in the small intestine by providing multiple niche signals. Recently, Paneth cells have become the focal point of investigations defining the mechanisms underlying the epithelium-microbiome interactions and pathogenesis of chronic gut mucosal inflammation and bacterial infection. Function of Paneth cells is tightly regulated by numerous factors at different levels, while Paneth cell defects have been widely documented in various gut mucosal diseases in humans. The post-transcription events, specific change in mRNA stability and translation by RNA-binding proteins (RBPs) and noncoding RNAs (ncRNAs) are implicated in many aspects of gut mucosal physiology by modulating Paneth cell function. Deregulation of RBPs and ncRNAs and subsequent Paneth cell defects are identified as crucial elements of gut mucosal pathologies. Here, we overview the posttranscriptional regulation of Paneth cells by RBPs and ncRNAs, with a particular focus on the increasing evidence of RBP HuR and long ncRNA H19 in this process. We also discuss the involvement of Paneth cell dysfunction in altered susceptibility of the intestinal epithelium to chronic inflammation and bacterial infection following disrupted expression of HuR and H19.
Collapse
|
19
|
Xiao L, Rao JN, Wang JY. RNA-binding proteins and long noncoding RNAs in intestinal epithelial autophagy and barrier function. Tissue Barriers 2021; 9:1895648. [PMID: 33709880 DOI: 10.1080/21688370.2021.1895648] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The intestinal autophagy and barrier function are crucial for maintaining the epithelium homeostasis and tightly regulated through well-controlled mechanisms. RNA-binding proteins (RBPs) and long noncoding RNAs (lncRNAs) modulate gene expression at the posttranscription level and are intimately involved in different physiological processes and diverse human diseases. In this review, we first highlight the roles of several RBPs and lncRNAs in the regulation of intestinal epithelial autophagy and barrier function, particularly focusing on the emerging evidence of RBPs and lncRNAs in the control of mRNA stability and translation. We additionally discuss recent findings that the interactions between RBPs and lncRNAs alter the fate of their target transcripts and thus influence gut epithelium host defense in response to stressful environments. These exciting advances in understanding the posttranscriptional control of the epithelial autophagy and barrier function by RBPs and lncRNAs provide a strong rationale for developing new effective therapeutics based on targeting RBPs and/or lncRNAs to preserve the intestinal epithelial integrity in patients with critical illnesses.
Collapse
Affiliation(s)
- Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA.,Department of Pathology, Department of Veterans Affairs, USA
| |
Collapse
|
20
|
Hernández-Cáceres MP, Munoz L, Pradenas JM, Pena F, Lagos P, Aceiton P, Owen GI, Morselli E, Criollo A, Ravasio A, Bertocchi C. Mechanobiology of Autophagy: The Unexplored Side of Cancer. Front Oncol 2021; 11:632956. [PMID: 33718218 PMCID: PMC7952994 DOI: 10.3389/fonc.2021.632956] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Proper execution of cellular function, maintenance of cellular homeostasis and cell survival depend on functional integration of cellular processes and correct orchestration of cellular responses to stresses. Cancer transformation is a common negative consequence of mismanagement of coordinated response by the cell. In this scenario, by maintaining the balance among synthesis, degradation, and recycling of cytosolic components including proteins, lipids, and organelles the process of autophagy plays a central role. Several environmental stresses activate autophagy, among those hypoxia, DNA damage, inflammation, and metabolic challenges such as starvation. In addition to these chemical challenges, there is a requirement for cells to cope with mechanical stresses stemming from their microenvironment. Cells accomplish this task by activating an intrinsic mechanical response mediated by cytoskeleton active processes and through mechanosensitive protein complexes which interface the cells with their mechano-environment. Despite autophagy and cell mechanics being known to play crucial transforming roles during oncogenesis and malignant progression their interplay is largely overlooked. In this review, we highlight the role of physical forces in autophagy regulation and their potential implications in both physiological as well as pathological conditions. By taking a mechanical perspective, we wish to stimulate novel questions to further the investigation of the mechanical requirements of autophagy and appreciate the extent to which mechanical signals affect this process.
Collapse
Affiliation(s)
- Maria Paz Hernández-Cáceres
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Leslie Munoz
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Javiera M. Pradenas
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Pena
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Lagos
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Aceiton
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Gareth I. Owen
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
- Facultad De Odontología, Instituto De Investigación En Ciencias Odontológicas (ICOD), Universidad De Chile, Santiago, Chile
| | - Andrea Ravasio
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| |
Collapse
|
21
|
Ni Y, Zhao Y, Ma L, Wang Z, Ni L, Hu L, Fu Z. Pharmacological activation of REV-ERBα improves nonalcoholic steatohepatitis by regulating intestinal permeability. Metabolism 2021; 114:154409. [PMID: 33096076 DOI: 10.1016/j.metabol.2020.154409] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/05/2020] [Accepted: 10/18/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES The gut-liver axis plays an important role in the pathogenesis of nonalcoholic steatohepatitis (NASH), and increased intestinal permeability causes transfer of endotoxin to the liver, which activates the immune response, ultimately leading to hepatic inflammation. Nuclear receptor Rev-erbα is a critical regulator of circadian rhythm, cellular metabolism, and inflammatory responses. However, the role and mechanism of Rev-erbα in gut barrier function and NASH remain unclear. In the present study, we investigated the involvement of Rev-erbα in the regulation of intestinal permeability and the treatment of NASH. METHODS AND RESULTS The expression of tight junction-related genes and Rev-erbs decreased in the jejunum, ileum and colon of mice with high cholesterol, high fat diet (CL)-induced NASH. Chromatin immunoprecipitation analysis indicated that REV-ERBα directly bound to the promoters of tight junction genes to regulate intestinal permeability. Pharmacological activation of REV-ERBα by SR9009 protected against lipopolysaccharide-induced increased intestinal permeability both in vitro and in vivo, and these effects were associated with the activation of autophagy and decreased apoptotic signaling of epithelial cells. In addition, the chronopharmacological effects of SR9009 were more potent at Zeitgeber time 0 (ZT0) than at ZT12, which was contrary to the rhythm of Rev-erbs in the gastrointestinal tract. The administration of SR9009 attenuated hepatic lipid accumulation, insulin resistance, inflammation, and fibrosis in mice with CL diet-induced NASH, which might be partly attributed to the enhancement of intestinal barrier function. CONCLUSION Chronopharmacological activation of REV-ERBα might be a potential strategy to treat intestinal barrier dysfunction-related disorders and NASH.
Collapse
Affiliation(s)
- Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yufeng Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Lingyan Ma
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhe Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Liyang Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Luting Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
22
|
Yang T, Heydarian M, Kozjak-Pavlovic V, Urban M, Harbottle RP, Rudel T. Folliculin Controls the Intracellular Survival and Trans-Epithelial Passage of Neisseria gonorrhoeae. Front Cell Infect Microbiol 2020; 10:422. [PMID: 33014885 PMCID: PMC7499807 DOI: 10.3389/fcimb.2020.00422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/08/2020] [Indexed: 12/18/2022] Open
Abstract
Neisseria gonorrhoeae, a Gram-negative obligate human pathogenic bacterium, infects human epithelial cells and causes sexually transmitted diseases. Emerging multi-antibiotic resistant gonococci and increasing numbers of infections complicate the treatment of infected patients. Here, we used an shRNA library screen and next-generation sequencing to identify factors involved in epithelial cell infection. Folliculin (FLCN), a 64 kDa protein with a tumor repressor function was identified as a novel host factor important for N. gonorrhoeae survival after uptake. We further determined that FLCN did not affect N. gonorrhoeae adherence and invasion but was essential for its survival in the cells by modulating autophagy. In addition, FLCN was also required to maintain cell to cell contacts in the epithelial layer. In an infection model with polarized cells, FLCN inhibited the polarized localization of E-cadherin and the transcytosis of gonococci across polarized epithelial cells. In conclusion, we demonstrate here the connection between FLCN and bacterial infection and in particular the role of FLCN in the intracellular survival and transcytosis of gonococci across polarized epithelial cell layers.
Collapse
Affiliation(s)
- Tao Yang
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | | | | | - Manuela Urban
- DNA Vector Lab, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | | | - Thomas Rudel
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Zheng J, Zeng M, Nian JB, Zeng LY, Fu Z, Huang QJ, Wei X. The CXCR4/miR-125b/FoxP3 axis regulates the function of the epithelial barrier via autophagy in allergic rhinitis. Am J Transl Res 2020; 12:2570-2584. [PMID: 32655791 PMCID: PMC7344073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 05/18/2020] [Indexed: 06/11/2023]
Abstract
An impaired epithelial barrier is often observed in allergic rhinitis (AR), which facilitates the infiltration of allergens. The aim of this study was to investigate the role of autophagy in the impaired epithelial barrier in AR and the related signalling pathways. A human nasal epithelial cell line was treated with dust mite allergen (Derp1). Autophagy was evaluated by GFP-LC3 adenovirus transfection and measurement of autophagy-related proteins. Epithelial barrier function was evaluated by measuring tight junction protein expression, transepithelial electrical resistance, and fluorescein isothiocyanate-dextran (FD4) permeability. Next, miR-125b inhibitor, miR-125b mimics, shRNA targeting FoxP3, pcDNA3.1 expressing FoxP3, and inhibitor of C-X-C motif chemokine receptor type 4 (CXCR4) were used to investigate the roles of miR-125b, FoxP3, and CXCR4 in epithelial cell autophagy and epithelial barrier function. An in vivo AR model was generated by exposing rat nasal mucosa to an allergen. Derp1 exposure enhanced autophagy and impaired the epithelial barrier in epithelial cells. Upregulation of miR-125b expression led to enhanced autophagy and impaired epithelial barrier through inhibition of FoxP3. Derp1 exposure increased miR-125b expression by increasing the expression and activation of CXCR4, which downregulated FoxP3 expression and led to enhanced autophagy and an impaired epithelial barrier. In vivo analysis confirmed the role of the CXCR4/miR-125b/FoxP3 axis in the impaired epithelial barrier in AR. This study demonstrates that the CXCR4/miR-125b/FoxP3 axis may participate in the pathogenesis of AR by regulating autophagy in epithelial cells and dysfunction of the epithelial barrier.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan Province, P. R. China
| | - Min Zeng
- Medical Center, Hainan Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan Province, P. R. China
| | - Jia-Bin Nian
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan Province, P. R. China
| | - Lian-Ya Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan Province, P. R. China
| | - Zhi Fu
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan Province, P. R. China
| | - Qiu-Ju Huang
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan Province, P. R. China
| | - Xin Wei
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan Province, P. R. China
| |
Collapse
|
24
|
Nighot P, Ma T. Endocytosis of Intestinal Tight Junction Proteins: In Time and Space. Inflamm Bowel Dis 2020; 27:283-290. [PMID: 32497180 PMCID: PMC7813749 DOI: 10.1093/ibd/izaa141] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Indexed: 12/12/2022]
Abstract
Eukaryotic cells take up macromolecules and particles from the surrounding milieu and also internalize membrane proteins via a precise process of endocytosis. The role of endocytosis in diverse physiological processes such as cell adhesion, cell signaling, tissue remodeling, and healing is well recognized. The epithelial tight junctions (TJs), present at the apical lateral membrane, play a key role in cell adhesion and regulation of paracellular pathway. These vital functions of the TJ are achieved through the dynamic regulation of the presence of pore and barrier-forming proteins within the TJ complex on the plasma membrane. In response to various intracellular and extracellular clues, the TJ complexes are actively regulated by intracellular trafficking. The intracellular trafficking consists of endocytosis and recycling cargos to the plasma membrane or targeting them to the lysosomes for degradation. Increased intestinal TJ permeability is a pathological factor in inflammatory bowel disease (IBD), and the TJ permeability could be increased due to the altered endocytosis or recycling of TJ proteins. This review discusses the current information on endocytosis of intestinal epithelial TJ proteins. The knowledge of the endocytic regulation of the epithelial TJ barrier will provide further understanding of pathogenesis and potential targets for IBD and a wide variety of human disease conditions.
Collapse
Affiliation(s)
- Prashant Nighot
- Department of Medicine, College of Medicine, Penn State University, Hershey, PA, USA,Address correspondence to: Prashant Nighot, Department of Medicine, College of Medicine, Pennsylvania State University, 500 University Drive, Room C5814B, Hershey, PA, 17033, USA. E-mail:
| | - Thomas Ma
- Department of Medicine, College of Medicine, Penn State University, Hershey, PA, USA
| |
Collapse
|
25
|
He B, Wang X, Yang C, Zhu J, Jin Y, Fu Z. The regulation of autophagy in the pesticide-induced toxicity: Angel or demon? CHEMOSPHERE 2020; 242:125138. [PMID: 31670000 DOI: 10.1016/j.chemosphere.2019.125138] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 05/20/2023]
Abstract
Pesticides have become an essential tool for pest kill, weed control and microbiome inhibition for both agricultural and domestic use. However, with the massive use, pesticides can exist in soil, air and water, and sometimes even accumulate in the human or other mammals through food chains. Lots of researches have proven that pesticides possess toxicity to mammals on endocrine, neural and immune systems. Autophagy, as a conservative intracellular process, which is activated by stress-related signals, plays a pivotal role, either "angle" or "demon", in regulation of cell fate and function. Recent evidences in researches elucidated a strong link between the autophagy and the toxicity of pesticides. In this review, we summarized the previous researches which focus on the autophagy regulation in the pesticides-induced toxicity, and hope that this work can help us to discover a potential strategy for the treatment of the disease caused by pesticides.
Collapse
Affiliation(s)
- Bingnan He
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Xia Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Chunlei Yang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Jianbo Zhu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China.
| |
Collapse
|
26
|
Somparn P, Boonkrai C, Charngkaew K, Chomanee N, Hodge KG, Fenton RA, Pisitkun T, Khositseth S. Bilateral ureteral obstruction is rapidly accompanied by ER stress and activation of autophagic degradation of IMCD proteins, including AQP2. Am J Physiol Renal Physiol 2020; 318:F135-F147. [PMID: 31736351 PMCID: PMC7054639 DOI: 10.1152/ajprenal.00113.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
After the release of bilateral ureteral obstruction (BUO), postobstructive diuresis from an impaired urine concentration mechanism is associated with reduced aquaporin 2 (AQP2) abundance in the inner medullary collecting duct (IMCD). However, the underlying molecular mechanism of this AQP2 reduction is incompletely understood. To elucidate the mechanisms responsible for this phenomenon, we studied molecular changes in IMCDs isolated from rats with 4-h BUO or sham operation at the early onset of AQP2 downregulation using mass spectrometry-based proteomic analysis. Two-hundred fifteen proteins had significant changes in abundances, with 65% of them downregulated in the IMCD of 4-h BUO rats compared with sham rats. Bioinformatic analysis revealed that significantly changed proteins were associated with functional Gene Ontology terms, including “cell-cell adhesion,” “cell-cell adherens junction,” “mitochondrial inner membrane,” “endoplasmic reticulum chaperone complex,” and the KEGG pathway of glycolysis/gluconeogenesis. Targeted liquid chromatography-tandem mass spectrometry or immunoblot analysis confirmed the changes in 19 proteins representative of each predominant cluster, including AQP2. Electron microscopy demonstrated disrupted tight junctions, disorganized adherens junctions, swollen mitochondria, enlargement of the endoplasmic reticulum lumen, and numerous autophagosomes/lysosomes in the IMCD of rats with 4-h BUO. AQP2 and seven proteins chosen as representative of the significantly altered clusters had a significant increase in immunofluorescence-based colocalization with autophagosomes/lysosomes. Immunogold electron microscopy confirmed colocalization of AQP2 with the autophagosome marker microtubule-associated protein 1A/1B-light chain 3 and the lysosomal marker cathepsin D in IMCD cells of rats with 4-h BUO. We conclude that enhanced autophagic degradation of AQP2 and other critical proteins, as well as endoplasmic reticulum stress in the IMCD, are initiated shortly after BUO.
Collapse
Affiliation(s)
- Poorichaya Somparn
- Center of Excellent in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Translational Research in Inflammation and Immunology Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chatikorn Boonkrai
- Center of Excellent in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Komgrid Charngkaew
- Department of Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand
| | - Nusara Chomanee
- Department of Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand
| | - Kenneth G Hodge
- Center of Excellent in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Robert A Fenton
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport, Aarhus University, Aarhus, Denmark
| | - Trairak Pisitkun
- Center of Excellent in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Sookkasem Khositseth
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| |
Collapse
|
27
|
Quantitative Proteome Analysis of Atg5-Deficient Mouse Embryonic Fibroblasts Reveals the Range of the Autophagy-Modulated Basal Cellular Proteome. mSystems 2019; 4:4/6/e00481-19. [PMID: 31690592 PMCID: PMC6832020 DOI: 10.1128/msystems.00481-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Autophagy performs housekeeping functions for cells and maintains a functional mode by degrading damaged proteins and organelles and providing energy under starvation conditions. The process is tightly regulated by the evolutionarily conserved Atg genes, of which Atg5 is one such crucial mediator. Here, we have done a comprehensive quantitative proteome analysis of mouse embryonic fibroblasts that lack a functional autophagy pathway (Atg5 knockout). We observe that 14% of the identified cellular proteome is remodeled, and several proteins distributed across diverse cellular processes with functions in signaling, cell adhesion, development, and immunity show either higher or lower levels under autophagy-deficient conditions. These cells have lower levels of crucial immune proteins that are required to mount a protective inflammatory response. This study will serve as a valuable resource to determine the role of autophagy in modulating specific protein levels in cells. Basal autophagy is crucial for maintenance of cellular homeostasis. ATG5 is an essential protein for autophagosome formation, and its depletion has been extensively used as a tool to disrupt autophagy. Here, we characterize the impact of Atg5 deficiency on the cellular proteome of mouse embryonic fibroblasts (MEFs). Using a tandem mass tagging (TMT)-based quantitative proteomics analysis, we observe that 14% of identified proteins show dysregulated levels in atg5−/− MEFs. These proteins were distributed across diverse biological processes, such as cell adhesion, development, differentiation, transport, metabolism, and immune responses. Several of the upregulated proteins were receptors involved in transforming growth factor β (TGF-β) signaling, JAK-STAT signaling, junction adhesion, and interferon/cytokine-receptor interactions and were validated as autophagy substrates. Nearly equal numbers of proteins, including several lysosomal proteins and enzymes, were downregulated, suggesting a complex role of autophagy/ATG5 in regulating their levels. The atg5−/− MEFs had lower levels of key immune sensors and effectors, including Toll-like receptor 2 (TLR2), interferon regulatory factor 3 (IRF3), IRF7, MLKL, and STAT1/3/5/6, which were restored by reexpression of ATG5. While these cells could efficiently mount a type I interferon response to the double-stranded RNA (dsRNA) mimic poly(I·C), they were compromised in their inflammatory response to the bacterial pathogen-associated molecular patterns (PAMPs) lipopolysaccharide (LPS) and Pam3CSK4. Transcriptional activation and secretion of interleukin-6 (IL-6) in these cells could be recovered by ATG5 expression, supporting the role of autophagy in the TLR2-induced inflammatory response. This study provides a key resource for understanding the effect of autophagy/ATG5 deficiency on the fibroblast proteome. IMPORTANCE Autophagy performs housekeeping functions for cells and maintains a functional mode by degrading damaged proteins and organelles and providing energy under starvation conditions. The process is tightly regulated by the evolutionarily conserved Atg genes, of which Atg5 is one such crucial mediator. Here, we have done a comprehensive quantitative proteome analysis of mouse embryonic fibroblasts that lack a functional autophagy pathway (Atg5 knockout). We observe that 14% of the identified cellular proteome is remodeled, and several proteins distributed across diverse cellular processes with functions in signaling, cell adhesion, development, and immunity show either higher or lower levels under autophagy-deficient conditions. These cells have lower levels of crucial immune proteins that are required to mount a protective inflammatory response. This study will serve as a valuable resource to determine the role of autophagy in modulating specific protein levels in cells. Author Video: An author video summary of this article is available.
Collapse
|
28
|
Hansson E, Skiöldebrand E. Low-grade inflammation causes gap junction-coupled cell dysfunction throughout the body, which can lead to the spread of systemic inflammation. Scand J Pain 2019; 19:639-649. [PMID: 31251727 DOI: 10.1515/sjpain-2019-0061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/21/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS Gap junction-coupled cells form networks in different organs in the body. These networks can be affected by inflammatory stimuli and become dysregulated. Cell signaling is also changed through connexin-linked gap junctions. This alteration affects the surrounding cells and extracellular matrix in organs. These changes can cause the spread of inflammatory substances, thus affecting other network-linked cells in other organs in the body, which can give rise to systemic inflammation, which in turn can lead to pain that can turn into chronic. METHODS This is a review based on literature search and our own research data of inflammatory stimuli that can affect different organs and particularly gap-junction-coupled cells throughout the body. CONCLUSIONS A remaining question is which cell type or tissue is first affected by inflammatory stimuli. Can endotoxin exposure through the air, water and body start the process and are mast cells the first target cells that have the capacity to alter the physiological status of gap junction-coupled cells, thereby causing breakdown of different barrier systems? IMPLICATIONS Is it possible to address the right cellular and biochemical parameters and restore inflammatory systems to a normal physiological level by therapeutic strategies?
Collapse
Affiliation(s)
- Elisabeth Hansson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 3rd Floor, SE 413 45 Gothenburg, Sweden, Phone: +46-31-786 3363
| | - Eva Skiöldebrand
- Section of Pathology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden.,Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
29
|
Feng Y, Huang Y, Wang Y, Wang P, Song H, Wang F. Antibiotics induced intestinal tight junction barrier dysfunction is associated with microbiota dysbiosis, activated NLRP3 inflammasome and autophagy. PLoS One 2019; 14:e0218384. [PMID: 31211803 PMCID: PMC6581431 DOI: 10.1371/journal.pone.0218384] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
Tight junction barrier is critical to intestinal homeostasis. Applying antibiotics to treat infections is common in clinical practice, which may affect intestinal microbiota. Intestinal microbiota dysbiosis is involved in the occurrence of some gastrointestinal diseases. Therefore, this study was aimed to investigate the influence of antibiotics on intestinal tight junction barrier and the possible underlying mechanisms. Healthy adult female C57BL/6 mice were treated with a broad-spectrum antibiotic cocktail for 14 days. 16S rDNA Illumina sequencing and headspace gas chromatography-mass spectrometry (HS-GC/MS) were respectively used to analyze microbial community and to detect short-chain fatty acids (SCFAs) contents. In vivo intestinal paracellular permeability to fluorescein isothiocyanate-dextran (FITC-dextran) was measured. Protein expression was determined by immunoblotting. Immunofluoresence was applied to observe the distributions of ZO-1, LC3B and ASC. Antibiotics remarkably altered intestinal microbiota composition in healthy mice, accompanying reduced SCFAs' concentrations. In addition, the intestinal tight junction barrier was disrupted by antibiotic treatment, as evidenced by increased intestinal paracellular permeability to FITC-dextran, decreased tight junction protein expressions, and disrupted ZO-1 morphology. Furthermore, NLRP3 inflammasome and autophagy were activated by antibiotic treatment. In conclusion, intestinal epithelial tight junction barrier dysfunction induced by antibiotics is associated with intestinal microbiota dysbiosis, activated NLRP3 inflammasome and autophagy in mice.
Collapse
Affiliation(s)
- Yanhai Feng
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yalan Huang
- Department of Military Nursing, School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu Wang
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pei Wang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Huapei Song
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengjun Wang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
30
|
Wu Y, Tang L, Wang B, Sun Q, Zhao P, Li W. The role of autophagy in maintaining intestinal mucosal barrier. J Cell Physiol 2019; 234:19406-19419. [PMID: 31020664 DOI: 10.1002/jcp.28722] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/23/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
The intestinal mucosal barrier is the first line to defense against luminal content penetration and performs numerous biological functions. The intestinal epithelium contains a huge surface that is lined by a monolayer of intestinal epithelial cells (IECs). IECs are dominant mediators in maintaining intestinal homeostasis that drive diverse functions including nutrient absorption, physical segregation, secretion of antibacterial peptides, and modulation of immune responses. Autophagy is a cellular self-protection mechanism in response to various stresses, and accumulating studies have revealed its importance in participating physiological processes of IECs. The regulatory effects of autophagy depend on the specific IEC types. This review aims to elucidate the myriad roles of autophagy in regulating the functions of different IECs (stem cells, enterocytes, goblet cells, and Paneth cells), and present the progress of autophagy-targeting therapy in intestinal diseases. Understanding the involved mechanisms can provide new preventive and therapeutic strategies for gastrointestinal dysfunction and diseases.
Collapse
Affiliation(s)
- Yanping Wu
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Li Tang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Baikui Wang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qiming Sun
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pengwei Zhao
- Department of Biochemistry, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
31
|
Retnakumar SV, Muller S. Pharmacological Autophagy Regulators as Therapeutic Agents for Inflammatory Bowel Diseases. Trends Mol Med 2019; 25:516-537. [PMID: 30952481 DOI: 10.1016/j.molmed.2019.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/03/2019] [Accepted: 03/06/2019] [Indexed: 12/12/2022]
Abstract
The arsenal of effective molecules to treat patients with chronic inflammatory bowel diseases (IBDs) remains limited. These remitting-relapsing diseases have become a global health issue and new therapeutic strategies are eagerly awaited to regulate the course of these disorders. Since the association between autophagy-related gene polymorphism and an increased risk of Crohn's disease (CD) has been discovered, a new domain of investigation has emerged, focused on the intracellular degradation system, with the objective of generating new medicines that are safer and more targeted. This review summarizes the drugs administered to IBD patients and describes recently emerged therapeutic agents. We compile evidence on the contribution of autophagy to IBD pathogenesis, give an overview of pharmacological autophagy regulators in animal models of colitis, and propose novel therapeutic avenues based on autophagy components.
Collapse
Affiliation(s)
- Sruthi Vijaya Retnakumar
- CNRS-University of Strasbourg, Biotechnology and Cell signaling, Institut de Science et d'ingénierie Supramoléculaire, 67000 Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell signaling, Institut de Science et d'ingénierie Supramoléculaire, 67000 Strasbourg, France; University of Strasbourg Institute for Advanced Study, 67000 Strasbourg, France.
| |
Collapse
|
32
|
Wong M, Ganapathy AS, Suchanec E, Laidler L, Ma T, Nighot P. Intestinal epithelial tight junction barrier regulation by autophagy-related protein ATG6/beclin 1. Am J Physiol Cell Physiol 2019; 316:C753-C765. [PMID: 30892937 DOI: 10.1152/ajpcell.00246.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A defective tight junction (TJ) barrier is a key pathogenic factor for inflammatory bowel disease. Previously, we have shown that autophagy, a cell survival mechanism, enhances intestinal epithelial TJ barrier function. Autophagy-related protein-6 (ATG6/beclin 1), a key protein in the autophagy pathway, also plays a role in the endocytic pathway. The constitutive role of beclin 1 in the intestinal TJ barrier is not known. In Caco-2 cells, beclin 1 was found to be coimmunoprecipitated with the TJ protein occludin and colocalized with occludin on the membrane. Treatment of Caco-2 cells with beclin 1 peptide [transactivating regulatory protein (Tat)-beclin 1] reduced TJ barrier function. Activation of beclin 1 increased occludin endocytosis and reduced total occludin protein level. In contrast, beclin 1 siRNA transfection enhanced Caco-2 TJ barrier function. In pharmacologic and genetic autophagy inhibition studies, the constitutive function of beclin 1 in the TJ barrier was found to be autophagy independent. However, de novo induction of autophagy with starvation or rapamycin prevented Tat-beclin 1-induced increase in TJ permeability and reduction in occludin level. Induction of autophagy also resulted in reduced beclin 1-occludin association. In mouse colon, beclin 1 colocalized with occludin on the epithelial membrane. Perfusion of mouse colon with beclin 1 peptide caused an increase in colonic TJ permeability that was prevented by in vivo induction of autophagy. These findings show that beclin 1 plays a constitutive, autophagy-independent role in the regulation of intestinal TJ barrier function via endocytosis of occludin. Autophagy terminates constitutive beclin 1 function in the TJ barrier and enhances the TJ barrier.
Collapse
Affiliation(s)
- Morgan Wong
- Department of Internal Medicine, University of New Mexico School of Medicine , Albuquerque, New Mexico
| | | | - Eric Suchanec
- Department of Medicine, College of Medicine, Pennsylvania State University , Hershey, Pennsylvania
| | - Laura Laidler
- Department of Internal Medicine, University of New Mexico School of Medicine , Albuquerque, New Mexico
| | - Thomas Ma
- Department of Medicine, College of Medicine, Pennsylvania State University , Hershey, Pennsylvania
| | - Prashant Nighot
- Department of Medicine, College of Medicine, Pennsylvania State University , Hershey, Pennsylvania
| |
Collapse
|
33
|
Khanna S, Domingo-Fernández D, Iyappan A, Emon MA, Hofmann-Apitius M, Fröhlich H. Using Multi-Scale Genetic, Neuroimaging and Clinical Data for Predicting Alzheimer's Disease and Reconstruction of Relevant Biological Mechanisms. Sci Rep 2018; 8:11173. [PMID: 30042519 PMCID: PMC6057884 DOI: 10.1038/s41598-018-29433-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 06/29/2018] [Indexed: 01/02/2023] Open
Abstract
Alzheimer's Disease (AD) is among the most frequent neuro-degenerative diseases. Early diagnosis is essential for successful disease management and chance to attenuate symptoms by disease modifying drugs. In the past, a number of cerebrospinal fluid (CSF), plasma and neuro-imaging based biomarkers have been proposed. Still, in current clinical practice, AD diagnosis cannot be made until the patient shows clear signs of cognitive decline, which can partially be attributed to the multi-factorial nature of AD. In this work, we integrated genotype information, neuro-imaging as well as clinical data (including neuro-psychological measures) from ~900 normal and mild cognitively impaired (MCI) individuals and developed a highly accurate machine learning model to predict the time until AD is diagnosed. We performed an in-depth investigation of the relevant baseline characteristics that contributed to the AD risk prediction. More specifically, we used Bayesian Networks to uncover the interplay across biological scales between neuro-psychological assessment scores, single genetic variants, pathways and neuro-imaging related features. Together with information extracted from the literature, this allowed us to partially reconstruct biological mechanisms that could play a role in the conversion of normal/MCI into AD pathology. This in turn may open the door to novel therapeutic options in the future.
Collapse
Affiliation(s)
- Shashank Khanna
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Anandhi Iyappan
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Mohammad Asif Emon
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, Sankt Augustin, 53754, Germany.,Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany
| | - Holger Fröhlich
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53113, Bonn, Germany. .,UCB Biosciences GmbH, Alfred-Nobel Str. 10, 40789, Monheim, Germany.
| |
Collapse
|
34
|
Lu Y, An J, Liu Y, Ren L, Zhang L. MMP9 is involved in HO-1-mediated upregulation of apical junctional complex in Caco-2 cells under oxygen-glucose deprivation. Biochem Biophys Res Commun 2018; 498:125-131. [PMID: 29425821 DOI: 10.1016/j.bbrc.2018.02.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/05/2018] [Indexed: 12/31/2022]
Abstract
Ischemia reperfusion injury is a critical factor in the recovery process after intestine trauma and the functional restoration of intestinal reconstruction. This study was the first to explore the expression of apical junctional complex (AJC) induced by heme oxygenase-1 (HO-1) in Caco-2 cells in oxygen-glucose deprivation (OGD) models. Here we showed that HO-1 was upregulated after OGD. Notably, activation of HO-1 largely enhanced the expression of AJC proteins including Claudin-4, E-cadherin and β-catenin in Caco-2 cells, but decreased the expression of matrix metalloproteinase 9 (MMP9). Knockdown of HO-1 attenuated the OGD-induced overexpression of AJC proteins but promoted the expression of MMP9. Interestingly, inhibition of MMP9 further enhanced AJC expression. These results suggest that HO-1 is involved in OGD-evoked upregulation of AJC proteins, which is partly mediated by MMP9 pathway. High expression of HO-1 may play an important role in the pathophysiological process of ischemia reperfusion injury and has potential clinical value for the treatment of intestine related diseases.
Collapse
Affiliation(s)
- Yongqu Lu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Jie An
- Department of Pharmacology, Shandong University School of Medicine, Jinan, 250012, Shandong, China
| | - Yulin Liu
- Department of Gastrointestinal Surgery, QianFoShan Hospital Affiliated to Shandong University, Jinan, 250014, Shandong, China
| | - Lehao Ren
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | - Li Zhang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|