1
|
Shu T, Zhang J, Hu R, Zhou F, Li H, Liu J, Fan Y, Li X, Ding P. Qi Huang Fang improves intestinal barrier function and intestinal microbes in septic mice through NLRP3 inflammasome-mediated cellular pyroptosis. Transpl Immunol 2024; 85:102072. [PMID: 38857634 DOI: 10.1016/j.trim.2024.102072] [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: 12/08/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVE Sepsis has a high incidence, morbidity, and mortality rate and is a great threat to human safety. Gut health plays an important role in sepsis development. Qi Huang Fang (QHF) contains astragalus, rhubarb, zhishi, and atractylodes. It is used to treat syndromes of obstructive qi and deficiency of righteousness. This study aimed to investigate whether QHF improves intestinal barrier function and microorganisms in mice through NLRP3 inflammatory vesicle-mediated cellular focal death. METHODS A mouse model of sepsis was constructed by cecal ligation and puncture (CLP) of specific pathogen-free (SPF)-grade C57BL/6 mice after continuous gavage of low, medium, and high doses of astragalus formula or probiotics for 4 weeks. Twenty-four hours postoperatively, the mechanism of action of QHF in alleviating septic intestinal dysfunction and restoring intestinal microecology, thereby alleviating intestinal injury, was evaluated by pathological observation, immunohistochemistry, western blotting, ELISA, and 16S rDNA high-throughput sequencing. RESULTS Different doses of QHF and probiotics ameliorated intestinal injury and reduced colonic apoptosis in mice to varying degrees (P < 0.05). Meanwhile, different doses of QHF and probiotics were able to reduce the serum levels of IL-6, IL-1β, and TNF-α (P < 0.05); down-regulate the protein expression of NLRP3, caspase-1, and caspase-11 (P < 0.05); and up-regulate the protein expression of zonula occluden-1 (ZO-1) and occludin (P < 0.05), which improved the intestinal barrier function in mice. In addition, QHF decreased the relative abundance of harmful bacteria (Firmicutes, Muribaculaceae, Campilobacterota, Helicobacter, and Alistipes) and increased the relative abundance of beneficial bacteria (Bacteroidetes and Actinobacteria) (P < 0.05). CONCLUSION QHF improves intestinal barrier function and gut microbiology in mice via NLRP3 inflammasome-mediated cellular pyroptosis.
Collapse
Affiliation(s)
- Tingting Shu
- Department of Intensive Care Unit, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Jun Zhang
- Department of Intensive Care Unit, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Ruiying Hu
- Department of Emergency Medicine, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Fang Zhou
- Department of Emergency Medicine, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Hanyong Li
- Department of Intensive Care Unit, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Jing Liu
- Department of Medical, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Yanbo Fan
- Department of Science and Education Section, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Xucheng Li
- Department of Emergency Medicine, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Peiwu Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
2
|
Lin Y, Wang X, Zhang K, Wang L, Zhang L, Yang J. Trajectories of acute gastrointestinal injury grade in critically Ill children. BMC Pediatr 2024; 24:470. [PMID: 39044193 PMCID: PMC11265350 DOI: 10.1186/s12887-024-04947-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
OBJECTIVE To investigate the characteristics of different Acute Gastrointestinal Injury (AGI) grading trajectories and examine their impact on prognosis in the Pediatric Intensive Care Unit (PICU). METHODS This retrospective cohort study was conducted at a large children's hospital in China. The children admitted to the PICU were included. AGI grade was assessed every other day during the initial nine days following PICU admission. RESULTS A total of 642 children were included, of which 364 children (56.7%) exhibited varying degrees of gastrointestinal dysfunction (AGI grade ≥ 2). Based on the patterns of AGI grading over time, six groups were identified: low-stable group, low-fluctuating group, medium-decreasing group, medium-increasing group, high-decreasing group, high-persistent group. The high-persistent group accounted for approximately 90% of all recorded deaths. Compared to low-stable group, both the medium-increasing and high-persistent groups exhibited positive correlations with length of stay in PICU (PICU LOS) and length of stay (LOS). Compared to low-stable group, the five groups exhibited a negative correlation with the percentage of energy received by enteral nutrition (EN), as well as the protein received by EN. CONCLUSION This study identified six distinct trajectory groups of AGI grade in critically ill children. The pattern of AGI grade trajectories over time were associated with EN delivery proportions and clinical outcomes.
Collapse
Affiliation(s)
- Ying Lin
- Department of Nutrition, Tianjin Children's Hospital, Tianjin University Children's Hospital, 225 Longyan Rd, Beichen Dist, Tianjin, China
| | - Xiaomin Wang
- Pediatric Intensive Care Unit, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Kai Zhang
- Department of Nutrition, Tianjin Children's Hospital, Tianjin University Children's Hospital, 225 Longyan Rd, Beichen Dist, Tianjin, China
| | - Lijing Wang
- Pediatric Intensive Care Unit, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Liping Zhang
- Pediatric Intensive Care Unit, Tianjin Children's Hospital, Tianjin University Children's Hospital, Tianjin, China
| | - Junhong Yang
- Department of Nutrition, Tianjin Children's Hospital, Tianjin University Children's Hospital, 225 Longyan Rd, Beichen Dist, Tianjin, China.
| |
Collapse
|
3
|
Khan N, Kurnik-Łucka M, Latacz G, Gil K. Systematic-Narrative Hybrid Literature Review: Crosstalk between Gastrointestinal Renin-Angiotensin and Dopaminergic Systems in the Regulation of Intestinal Permeability by Tight Junctions. Int J Mol Sci 2024; 25:5566. [PMID: 38791603 PMCID: PMC11122119 DOI: 10.3390/ijms25105566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/12/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
In the first part of this article, the role of intestinal epithelial tight junctions (TJs), together with gastrointestinal dopaminergic and renin-angiotensin systems, are narratively reviewed to provide sufficient background. In the second part, the current experimental data on the interplay between gastrointestinal (GI) dopaminergic and renin-angiotensin systems in the regulation of intestinal epithelial permeability are reviewed in a systematic manner using the PRISMA methodology. Experimental data confirmed the copresence of DOPA decarboxylase (DDC) and angiotensin converting enzyme 2 (ACE2) in human and rodent enterocytes. The intestinal barrier structure and integrity can be altered by angiotensin (1-7) and dopamine (DA). Both renin-angiotensin and dopaminergic systems influence intestinal Na+/K+-ATPase activity, thus maintaining electrolyte and nutritional homeostasis. The colocalization of B0AT1 and ACE2 indicates the direct role of the renin-angiotensin system in amino acid absorption. Yet, more studies are needed to thoroughly define the structural and functional interaction between TJ-associated proteins and GI renin-angiotensin and dopaminergic systems.
Collapse
Affiliation(s)
- Nadia Khan
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 31-008 Krakow, Poland
| | - Magdalena Kurnik-Łucka
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
| | - Gniewomir Latacz
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 31-008 Krakow, Poland
| | - Krzysztof Gil
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
| |
Collapse
|
4
|
Lin Y, Wang X, Li L, Gou Y, Zhang L, Wang L, Yang J. Nomogram to predict feeding intolerance in critically ill children. Eur J Pediatr 2023; 182:5293-5302. [PMID: 37723295 DOI: 10.1007/s00431-023-05205-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/28/2023] [Accepted: 09/13/2023] [Indexed: 09/20/2023]
Abstract
Feed intolerance (FI) is significantly associated with poor prognosis in critically ill patients. This study aimed to understand the characteristics of children with FI and identify the factors predicting FI in critically ill children. This retrospective cohort study was conducted between January 2017 and June 2022 in the Pediatric Intensive Care Unit of a specialized children's hospital. Eighteen factors, including age, body mass index for age z-score (BAZ) < -2, paediatric index of mortality (PIM)3 score, Glasgow coma scale score, mechanical ventilation (MV), enteral nutrition delay, vasoactive drugs, sedatives, sepsis, heart disease, neurological disease, hypokalemia, arterial PH < 7.35, arterial partial pressure of oxygen (PaO2), blood glucose, hemoglobin, total protein, and albumin, were retrieved to predict FI. The outcome was FI during PICU stay. During the study period, a total of 854 children were included, of which 215 children developed FI. Six predictors of FI were selected: PIM3 score, MV, sepsis, hypokalemia, albumin, and PaO2. Multivariate logistic regression analysis showed that higher PIM3 score, MV, sepsis, hypokalemia, and lower PaO2 were independent risk factors for FI, whereas higher albumin was an independent protective factor for FI. The C-index of the predictive nomogram of 0.943 was confirmed at internal validation to be 0.940, indicating a good predictive value of the model. Decision curve analysis shows good clinical applicability of the nomogram in predicting FI. Conclusion: The nomogram was verified to have a good prediction performance based on discrimination, calibration, and clinical decision analysis. What is Known: • Research has demonstrated that gastrointestinal (GI) dysfunction is not only a fundamental element of Multiple Organ Dysfunction Syndrome (MODS), but also the initiator of MODS. • Previous study has demonstrated a significant association between FI and poor prognosis in critically ill patients. What is New: • We excluded patients with primary gastrointestinal tract disease from our study, and we observed an incidence of FI of 25.2% in the Pediatric Intensive Care Unit (PICU). • Our study revealed that PIM3 score, MV, sepsis, hypokalemia, albumin, and PaO2 are significant predictors of FI.
Collapse
Affiliation(s)
- Ying Lin
- Department of Nutrition, Tianjin Children's Hospital /Tianjin University Children's Hospital, 225 Longyan Rd, Beichen Dist, Tianjin, China.
| | - Xiaomin Wang
- Pediatric Intensive Care Unit, Tianjin Children's Hospital /Tianjin University Children's Hospital, Tianjin, China
| | - Lingyan Li
- Department of Nutrition, Tianjin Children's Hospital /Tianjin University Children's Hospital, 225 Longyan Rd, Beichen Dist, Tianjin, China
| | - Yun Gou
- Department of Nutrition, Tianjin Children's Hospital /Tianjin University Children's Hospital, 225 Longyan Rd, Beichen Dist, Tianjin, China
| | - Liping Zhang
- Pediatric Intensive Care Unit, Tianjin Children's Hospital /Tianjin University Children's Hospital, Tianjin, China
| | - Lijing Wang
- Pediatric Intensive Care Unit, Tianjin Children's Hospital /Tianjin University Children's Hospital, Tianjin, China
| | - Junhong Yang
- Department of Nutrition, Tianjin Children's Hospital /Tianjin University Children's Hospital, 225 Longyan Rd, Beichen Dist, Tianjin, China
| |
Collapse
|
5
|
Jonusaite S, Oulhen N, Izumi Y, Furuse M, Yamamoto T, Sakamoto N, Wessel G, Heyland A. Identification of the genes encoding candidate septate junction components expressed during early development of the sea urchin, Strongylocentrotus purpuratus, and evidence of a role for Mesh in the formation of the gut barrier. Dev Biol 2023; 495:21-34. [PMID: 36587799 DOI: 10.1016/j.ydbio.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022]
Abstract
Septate junctions (SJs) evolved as cell-cell junctions that regulate the paracellular barrier and integrity of epithelia in invertebrates. Multiple morphological variants of SJs exist specific to different epithelia and/or phyla but the biological significance of varied SJ morphology is unclear because the knowledge of the SJ associated proteins and their functions in non-insect invertebrates remains largely unknown. Here we report cell-specific expression of nine candidate SJ genes in the early life stages of the sea urchin Strongylocentrotus purpuratus. By use of in situ RNA hybridization and single cell RNA-seq we found that the expression of selected genes encoding putatively SJ associated transmembrane and cytoplasmic scaffold molecules was dynamically regulated during epithelial development in the embryos and larvae with different epithelia expressing different cohorts of SJ genes. We focused a functional analysis on SpMesh, a homolog of the Drosophila smooth SJ component Mesh, which was highly enriched in the endodermal epithelium of the mid- and hindgut. Functional perturbation of SpMesh by both CRISPR/Cas9 mutagenesis and vivo morpholino-mediated knockdown shows that loss of SpMesh does not disrupt the formation of the gut epithelium during gastrulation. However, loss of SpMesh resulted in a severely reduced gut-paracellular barrier as quantitated by increased permeability to 3-5 kDa FITC-dextran. Together, these studies provide a first look at the molecular SJ physiology during the development of a marine organism and suggest a shared role for Mesh-homologous proteins in forming an intestinal barrier in invertebrates. Results have implications for consideration of the traits underlying species-specific sensitivity of marine larvae to climate driven ocean change.
Collapse
Affiliation(s)
- Sima Jonusaite
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Nathalie Oulhen
- Department of Molecular and Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, United States
| | - Yasushi Izumi
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan; Nagoya University Graduate School of Medicine, Aichi, 464-8601, Japan
| | - Takashi Yamamoto
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8526, Japan
| | - Naoaki Sakamoto
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8526, Japan
| | - Gary Wessel
- Department of Molecular and Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, United States
| | - Andreas Heyland
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
6
|
Wu H, Huang R, Fan J, Luo N, Yang X. Low potassium disrupt intestinal barrier and result in bacterial translocation. Lab Invest 2022; 20:309. [PMID: 35794599 PMCID: PMC9258207 DOI: 10.1186/s12967-022-03499-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/24/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Bacterial translocation was observed in critical illness and patients with chronic diseases such as liver cirrhosis and chronic kidney disease (CKD). Hypokalemia is a common complication in these diseases. Whether low potassium diet may increase intestinal permeability and result in bacterial translocation lack of evidence. The present study was aimed to investigate the potential effects of LK on intestinal permeability.
Methods
Grade 8-week-old male Bal B/C mice were randomly placed either on a normal potassium (NK) mouse chow or a low potassium (LK) diet for 28 days. Intestinal permeability and expression of tight junction proteins were compared between the two groups.
Results
Compared with the NK group, the mice in LK group had significantly lower serum potassium level, increased levels of plasmas endotoxin and plasma d-lactate. The bacterial translocation was higher and in occurred mainly in mesenteric lymph nodes (MLN), liver and spleen. The pathologic change of small intestine was obvious with thinner villus lamina propria, shorter crypt depth and thinner intestinal wall. Slight increases in the expression of proteins and mRNA levels of both claudin-1 and claudin-2 were observed in LK group.
Conclusions
Low potassium diet could increase intestinal permeability and thereby lead to bacterial translocation, which was suspected to result from impaired intestinal epithelial barrier and biological barrier.
Collapse
|
7
|
Ashour L. Roles of the ACE/Ang II/AT1R pathway, cytokine release, and alteration of tight junctions in COVID-19 pathogenesis. Tissue Barriers 2022; 11:2090792. [PMID: 35726726 PMCID: PMC10161962 DOI: 10.1080/21688370.2022.2090792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This paper shows how SARS-CoV-2 alters tight junctions (TJs) in human organs. The effect of SARS-CoV-2 on the ACE/Ang II/AT1R pathway and immune cells culminates in the release of numerous pro-inflammatory mediators, leading to the presence of certain symptoms in COVID-19, such as acute lung injury (ALI), pulmonary hypertension, and pulmonary fibrosis. Furthermore, the cytokines released alter different TJs components. The study shows how the irregular release of pro-inflammatory cytokines leads to claudin disruption in various tissues of the body, resulting in different symptoms, such as alveolar fibrosis, pulmonary edema, conjunctivitis, altered fertility in males, gastrointestinal symptoms, Covid toes, and others. SARS-CoV-2 also alters occludin expression in the endothelial and blood-testis barriers (BTB) resulting in edema and altered fertility. Viral disruption of JAM-A leads to activation of the RhoA GTPase, which leads to ALI. Taken together, these results define ACE/Ang II/AT1R pathway receptors and tight junctional components as potential therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Laith Ashour
- Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| |
Collapse
|
8
|
Liu X, Zhu H. Curcumin Improved Intestinal Epithelial Barrier Integrity by Up-Regulating ZO-1/Occludin/Claudin-1 in Septic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2884522. [PMID: 35711494 PMCID: PMC9197639 DOI: 10.1155/2022/2884522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 11/17/2022]
Abstract
Objective To investigate the protective effect and mechanism of curcumin on intestinal barrier function in rats with enterogenic sepsis. Methods Rats were divided into Sham group (Sham), Model group (Model), low-dose curcumin group (100 mg/kg), and high-dose curcumin group (200 mg/kg), with 10 rats in each group. Sepsis model was established in model group, low-dose curcumin group, and high-dose curcumin group. After drug intervention, hematoxylin-eosin (HE) staining was used to observe the histopathological changes of small intestine in each group. The levels of TNF-α, IL-1β, and IL-6 in serum and intestinal tissues of rats were determined by ELISA. The expression of ZO-1, occludin, and claudin-1 in ileum was detected by QRT-PCR and Western blot. Western blotting was used to detect the expression of ERK/JNK signaling pathway, NF-κB p65, apoptosis-related proteins Caspase-3, and TNF-α in rat intestinal tissues. Results HE staining showed that curcumin treatment reduced epithelial cell shedding, interstitial edema, and apoptosis. Compared with model group, DAO activity, serum intestinal fatty acid binding protein (I-FABP), TNF-α, IL-6, and IL-1β expression in curcumin group were decreased in a dose-dependent manner. Curcumin can upregulate the mRNA and protein expression levels of ZO-1, occludin, and claudin-1 in ileum of CLP-induced rats. In addition, curcumin inhibits NF-κB p65 activation and apoptosis by regulating ERK/JNK signaling pathway. Conclusion Curcumin can reduce inflammatory response and upregulate the expression of intestinal tight junction proteins ZO-1, occludin, and claudin-1 in rats with enterogenic sepsis, and protect intestinal barrier function.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Intensive Care Unit, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Hongquan Zhu
- Intensive Care Unit, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| |
Collapse
|
9
|
Hao W, Zhu X, Liu Z, Song Y, Wu S, Lu X, Yang J, Jin C. Resveratrol alleviates aluminum-induced intestinal barrier dysfunction in mice. ENVIRONMENTAL TOXICOLOGY 2022; 37:1373-1381. [PMID: 35156769 DOI: 10.1002/tox.23490] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/27/2022] [Accepted: 02/05/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Aluminum is mainly exposed to the general population through food and water, and is absorbed into the systemic circulation through intestine, which in turn damages the intestinal barrier. METHODS The mice model of subchronic exposure to aluminum chloride (AlCl3 ) was established via oral. Tail suspension test was used to detect depressive behavior. H&E staining was performed to assess pathological intestinal injury. Intestinal permeability was estimated by exogenous Evans blue content. The level of inflammatory cytokines and tight junction protein were assessed via ELISA and western blotting. Simultaneously, resveratrol (Rsv, an agonist of Sirt1) was evaluated the protective role against intestinal barrier injuries caused by aluminum exposure. RESULTS Our results showed that AlCl3 induced depressive-like behavior, intestinal pathological damage and intestinal barrier permeability, resulting in intestinal barrier dysfunction. Besides, aluminum induced the expression of inflammatory cytokines, which further triggered IRF8-MMP9-mediated downregulation of tight junction proteins including CLD1, OCLD and ZO-1. After Rsv treatment, SIRT1 expression was increased, depressive symptom was improved, pathological injury was reduced, inflammatory reaction was alleviated, and intestinal barrier function restored. CONCLUSION Our findings revealed that aluminum exposure induced intestinal barrier dysfunction by IRF8-MMP9 signaling pathway. Rsv alleviated these injuries via activating SIRT1.
Collapse
Affiliation(s)
- Wudi Hao
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Xiaoying Zhu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Ziyue Liu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Yushuai Song
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Shengwen Wu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Xiaobo Lu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Jinghua Yang
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Cuihong Jin
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
10
|
Hao W, Hao C, Wu C, Xu Y, Jin C. Aluminum induced intestinal dysfunction via mechanical, immune, chemical and biological barriers. CHEMOSPHERE 2022; 288:132556. [PMID: 34648793 DOI: 10.1016/j.chemosphere.2021.132556] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/18/2021] [Accepted: 10/10/2021] [Indexed: 06/13/2023]
Abstract
Aluminum is the most abundant metal element in the Earth's crust, which exists naturally in the form of aluminum compounds. Aluminum is mainly absorbed through the gastrointestinal tract, which varies with different aluminum compounds. During this process, aluminum could induce the disruption of intestinal mucosa barrier. However, its underlying mechanism has not been elucidated yet. Previous studies have reported that aluminum can firstly promote the apoptosis of intestinal epithelial cells, destroy the structure of tight-junction proteins, and increase the intestinal permeability, injuring the mechanical barrier of gut. Also, it can induce the activation of immune cells to secrete inflammatory factors, and trigger immune responses, interfering with immune barrier. Moreover, aluminum treatment can regulate intestinal composition and bio-enzyme activity, impairing the function of chemical barrier. In addition, aluminum accumulation can induce an imbalance of the intestinal flora, inhibit the growth of beneficial bacteria, and promote the proliferation of harmful bacteria, which ultimately disrupting biological barrier. Collectively, aluminum may do extensive damage to intestinal barrier function covering mechanical barrier, immune barrier, chemical barrier and biological barrier.
Collapse
Affiliation(s)
- Wudi Hao
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Chenyu Hao
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Chengrong Wu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Yuqing Xu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Cuihong Jin
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China.
| |
Collapse
|
11
|
Liu J, Dean DA. Gene Therapy for Acute Respiratory Distress Syndrome. Front Physiol 2022; 12:786255. [PMID: 35111077 PMCID: PMC8801611 DOI: 10.3389/fphys.2021.786255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating clinical syndrome that leads to acute respiratory failure and accounts for over 70,000 deaths per year in the United States alone, even prior to the COVID-19 pandemic. While its molecular details have been teased apart and its pathophysiology largely established over the past 30 years, relatively few pharmacological advances in treatment have been made based on this knowledge. Indeed, mortality remains very close to what it was 30 years ago. As an alternative to traditional pharmacological approaches, gene therapy offers a highly controlled and targeted strategy to treat the disease at the molecular level. Although there is no single gene or combination of genes responsible for ARDS, there are a number of genes that can be targeted for upregulation or downregulation that could alleviate many of the symptoms and address the underlying mechanisms of this syndrome. This review will focus on the pathophysiology of ARDS and how gene therapy has been used for prevention and treatment. Strategies for gene delivery to the lung, such as barriers encountered during gene transfer, specific classes of genes that have been targeted, and the outcomes of these approaches on ARDS pathogenesis and resolution will be discussed.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| | - David A. Dean
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
12
|
Heymans C, Delcorte O, Spourquet C, Villacorte-Tabelin M, Dupasquier S, Achouri Y, Mahibullah S, Lemoine P, Balda MS, Matter K, Pierreux CE. Spatio-temporal expression pattern and role of the tight junction protein MarvelD3 in pancreas development and function. Sci Rep 2021; 11:14519. [PMID: 34267243 PMCID: PMC8282860 DOI: 10.1038/s41598-021-93654-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/23/2021] [Indexed: 11/29/2022] Open
Abstract
Tight junction complexes are involved in the establishment and maintenance of cell polarity and the regulation of signalling pathways, controlling biological processes such as cell differentiation and cell proliferation. MarvelD3 is a tight junction protein expressed in adult epithelial and endothelial cells. In Xenopus laevis, MarvelD3 morphants present differentiation defects of several ectodermal derivatives. In vitro experiments further revealed that MarvelD3 couples tight junctions to the MEKK1-JNK pathway to regulate cell behaviour and survival. In this work, we found that MarvelD3 is expressed from early developmental stages in the exocrine and endocrine compartments of the pancreas, as well as in endothelial cells of this organ. We thoroughly characterized MarvelD3 expression pattern in developing pancreas and evaluated its function by genetic ablation. Surprisingly, inactivation of MarvelD3 in mice did not alter development and differentiation of the pancreatic tissue. Moreover, tight junction formation and organization, cell polarization, and activity of the JNK-pathway were not impacted by the deletion of MarvelD3.
Collapse
Affiliation(s)
| | - Ophélie Delcorte
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe, Belgium
| | | | - Mylah Villacorte-Tabelin
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe, Belgium
- PRISM, MSU-IIT, Iligan City, Philippines
| | | | | | - Siam Mahibullah
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe, Belgium
| | - Pascale Lemoine
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe, Belgium
| | | | | | | |
Collapse
|
13
|
Kryvenko V, Vagin O, Dada LA, Sznajder JI, Vadász I. Maturation of the Na,K-ATPase in the Endoplasmic Reticulum in Health and Disease. J Membr Biol 2021; 254:447-457. [PMID: 34114062 PMCID: PMC8192048 DOI: 10.1007/s00232-021-00184-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/08/2021] [Indexed: 12/11/2022]
Abstract
Abstract The Na,K-ATPase establishes the electrochemical gradient of cells by driving an active exchange of Na+ and K+ ions while consuming ATP. The minimal functional transporter consists of a catalytic α-subunit and a β-subunit with chaperon activity. The Na,K-ATPase also functions as a cell adhesion molecule and participates in various intracellular signaling pathways. The maturation and trafficking of the Na,K-ATPase include co- and post-translational processing of the enzyme in the endoplasmic reticulum (ER) and the Golgi apparatus and subsequent delivery to the plasma membrane (PM). The ER folding of the enzyme is considered as the rate-limiting step in the membrane delivery of the protein. It has been demonstrated that only assembled Na,K-ATPase α:β-complexes may exit the organelle, whereas unassembled, misfolded or unfolded subunits are retained in the ER and are subsequently degraded. Loss of function of the Na,K-ATPase has been associated with lung, heart, kidney and neurological disorders. Recently, it has been shown that ER dysfunction, in particular, alterations in the homeostasis of the organelle, as well as impaired ER-resident chaperone activity may impede folding of Na,K-ATPase subunits, thus decreasing the abundance and function of the enzyme at the PM. Here, we summarize our current understanding on maturation and subsequent processing of the Na,K-ATPase in the ER under physiological and pathophysiological conditions. Graphic Abstract ![]()
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Klinikstrasse 33, 35392, Giessen, Germany.,The Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - Olga Vagin
- Department of Physiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Laura A Dada
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jacob I Sznajder
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - István Vadász
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Klinikstrasse 33, 35392, Giessen, Germany. .,The Cardio-Pulmonary Institute (CPI), Giessen, Germany.
| |
Collapse
|
14
|
Kryvenko V, Vadász I. Molecular mechanisms of Na,K-ATPase dysregulation driving alveolar epithelial barrier failure in severe COVID-19. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1186-L1193. [PMID: 33689516 PMCID: PMC8238442 DOI: 10.1152/ajplung.00056.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A significant number of patients with coronavirus disease 2019 (COVID-19) develop acute respiratory distress syndrome (ARDS) that is associated with a poor outcome. The molecular mechanisms driving failure of the alveolar barrier upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection remain incompletely understood. The Na,K-ATPase is an adhesion molecule and a plasma membrane transporter that is critically required for proper alveolar epithelial function by both promoting barrier integrity and resolution of excess alveolar fluid, thus enabling appropriate gas exchange. However, numerous SARS-CoV-2-mediated and COVID-19-related signals directly or indirectly impair the function of the Na,K-ATPase, thereby potentially contributing to disease progression. In this Perspective, we highlight some of the putative mechanisms of SARS-CoV-2-driven dysfunction of the Na,K-ATPase, focusing on expression, maturation, and trafficking of the transporter. A therapeutic mean to selectively inhibit the maladaptive signals that impair the Na,K-ATPase upon SARS-CoV-2 infection might be effective in reestablishing the alveolar epithelial barrier and promoting alveolar fluid clearance and thus advantageous in patients with COVID-19-associated ARDS.
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,The Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,The Cardio-Pulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
15
|
Sassi A, Wang Y, Chassot A, Komarynets O, Roth I, Olivier V, Crambert G, Dizin E, Boscardin E, Hummler E, Feraille E. Interaction between Epithelial Sodium Channel γ-Subunit and Claudin-8 Modulates Paracellular Sodium Permeability in Renal Collecting Duct. J Am Soc Nephrol 2020; 31:1009-1023. [PMID: 32245797 DOI: 10.1681/asn.2019080790] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 02/15/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Water and solute transport across epithelia can occur via the transcellular or paracellular pathways. Tight junctions play a key role in mediating paracellular ion reabsorption in the kidney. In the renal collecting duct, which is a typical absorptive tight epithelium, coordination between transcellular sodium reabsorption and paracellular permeability may prevent the backflow of reabsorbed sodium to the tubular lumen along a steep electrochemical gradient. METHODS To investigate whether transcellular sodium transport controls tight-junction composition and paracellular permeability via modulating expression of the transmembrane protein claudin-8, we used cultured mouse cortical collecting duct cells to see how overexpression or silencing of epithelial sodium channel (ENaC) subunits and claudin-8 affect paracellular permeability. We also used conditional kidney tubule-specific knockout mice lacking ENaC subunits to assess the ENaC's effect on claudin-8 expression. RESULTS Overexpression or silencing of the ENaC γ-subunit was associated with parallel and specific changes in claudin-8 abundance. Increased claudin-8 abundance was associated with a reduction in paracellular permeability to sodium, whereas decreased claudin-8 abundance was associated with the opposite effect. Claudin-8 overexpression and silencing reproduced these functional effects on paracellular ion permeability. Conditional kidney tubule-specific ENaC γ-subunit knockout mice displayed decreased claudin-8 expression, confirming the cell culture experiments' findings. Importantly, ENaC β-subunit or α-subunit silencing or kidney tubule-specific β-ENaC or α-ENaC knockout mice did not alter claudin-8 abundance. CONCLUSIONS Our data reveal the specific coupling between ENaC γ-subunit and claudin-8 expression. This coupling may play an important role in preventing the backflow of reabsorbed solutes and water to the tubular lumen, as well as in coupling paracellular and transcellular sodium permeability.
Collapse
Affiliation(s)
- Ali Sassi
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Yubao Wang
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Alexandra Chassot
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Olga Komarynets
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland
| | - Isabelle Roth
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland
| | - Valérie Olivier
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Gilles Crambert
- Sorbonne University, Unité Mixte de Recherche (UMR) S1138, Cordeliers Research Center, Paris, France
| | - Eva Dizin
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland.,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| | - Emilie Boscardin
- National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Edith Hummler
- National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Eric Feraille
- Department of Cellular Physiology and Metabolism, University of Geneva, University Medical Center, Geneva, Switzerland .,National Center of Competence in Research Kidney Control of Homeostasis (Kidney.CH), Zurich, Switzerland
| |
Collapse
|
16
|
Abstract
Acute and chronic pancreatitises are gastrointestinal inflammatory diseases, the incidence of which is increasing worldwide. Most (~ 80%) acute pancreatitis (AP) patients have mild disease, and about 20% have severe disease, which causes multiple organ failure and has a high mortality rate. Chronic pancreatitis (CP) is characterized by chronic inflammation and destruction of normal pancreatic parenchyma, which leads to loss of exocrine and endocrine tissues. Patients with CP also have a higher incidence of pancreatic ductal adenocarcinoma. Although a number of factors are associated with the development and progression of AP and CP, the underlying mechanism is unclear. Adhesion molecules play important roles in cell migration, proliferation, and signal transduction, as well as in development and tissue repair. Loosening of cell-cell adhesion between pancreatic acinar cells and/or endothelial cells increases solute permeability, resulting in interstitial edema, which promotes inflammatory cell migration and disrupts tissue structure. Oxidative stress, which is one of the important pathogenesis of pancreatitis, leads to upregulation of adhesion molecules. Soluble adhesion molecules are reportedly involved in AP. In this review, we focus on the roles of tight junctions (occludin, tricellulin, claudin, junctional adhesion molecule, and zonula occludin), adherens junctions (E-cadherin and p120-, α-, and β-catenin), and other adhesion molecules (selectin and intercellular adhesion molecules) in the progression of AP and CP. Maintaining the normal function of adhesion molecules and preventing their abnormal activation maintain the structure of the pancreas and prevent the development of pancreatitis.
Collapse
Affiliation(s)
- Takeshi Sato
- 0000 0001 1033 6139grid.268441.dDepartment of Gastroenterology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004 Japan
| | - Wataru Shibata
- 0000 0001 1033 6139grid.268441.dDepartment of Gastroenterology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004 Japan ,0000 0001 1033 6139grid.268441.dDivision of Translational Research, Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004 Japan
| | - Shin Maeda
- 0000 0001 1033 6139grid.268441.dDepartment of Gastroenterology, Yokohama City University Graduate School of Medicine, Fukuura 3-9, Kanazawa-ku, Yokohama, Kanagawa 236-0004 Japan
| |
Collapse
|
17
|
TRPA1-dependent reversible opening of tight junction by natural compounds with an α,β-unsaturated moiety and capsaicin. Sci Rep 2018; 8:2251. [PMID: 29396565 PMCID: PMC5797179 DOI: 10.1038/s41598-018-20526-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/19/2018] [Indexed: 12/25/2022] Open
Abstract
The delivery of hydrophilic macromolecules runs into difficulties such as penetration of the cell membrane lipid bilayer. Our prior experiment demonstrated that capsaicin induces the reversible opening of tight junctions (TJs) and enhances the delivery of hydrophilic macromolecules through a paracellular route. Herein, we screened paracellular permeability enhancers other than capsaicin. As TJ opening by capsaicin is associated with Ca2+ influx, we first screened the compounds that induce Ca2+ influx in layered MDCK II cells, and then we determined the compounds' abilities to open TJs. Our results identified several natural compounds with α,β-unsaturated moiety. A structure-activity relationship (SAR) analysis and the results of pretreatment with reducing reagent DTT suggested the importance of α,β-unsaturated moiety. We also examined the underlying mechanisms, and our findings suggest that the actin reorganization seen in capsaicin treatment is important for the reversibility of TJ opening. Furthermore, our analyses revealed that TRPA1 is involved in the Ca2+ influx and TJ permeability increase not only by an α,β-unsaturated compound but also by capsaicin. Our results indicate that the α,β-unsaturated moiety can be a potent pharmacophore for TJ opening.
Collapse
|
18
|
Kong WG, Li SS, Chen XX, Huang YQ, Tang Y, Wu ZX. A study of the damage of the intestinal mucosa barrier structure and function of Ctenopharyngodon idella with Aeromonas hydrophila. FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:1223-1235. [PMID: 28425012 DOI: 10.1007/s10695-017-0366-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/16/2017] [Indexed: 06/07/2023]
Abstract
The aim of this study is to explore the effect of Aeromonas hydrophila on the intestinal mucosal barrier structure and intestinal permeability in grass carp (Ctenopharyngodon idella). Histopathological examinations showed that A. hydrophila induced severe intestinal lesions, including inflammatory cell infiltration and intestinal villus fusion and swelling. Messenger RNA (mRNA) expression of the inflammatory cytokines TNF-α, IL-1β, IL-8, IL-10 and MyD88 was significantly increased after infection with A. hydrophila. The permeability of intestinal mucosa was determined using Evans blue (EB) and D-lactic acid. The results indicated that the levels of EB and serum D-lactic acid were significantly increased after infection with A. hydrophila (p < 0.05). Our results also indicated that the intestinal mucosal barrier injury induced by A. hydrophila infection was closely associated with the expression of the tight junction (TJ) protein zonula occludens-1 (ZO-1), occludin, claudin b and claudin c as well as the activity of Na+, K+-ATPase and Ca2+, Mg2+-ATPase. Lower mRNA levels of occludin and lower Na+, K+-ATPase and Ca2+, Mg2+-ATPase activity in the intestines were observed after challenge. ZO-1 and claudin c were significantly increased 24 h after infection with A. hydrophila. The most interesting finding was that claudin b also significantly increased 24 h after challenge and then decreased to lower levels at 72, 120 and 168 h post-infection compared to the PBS-treated control group. The results demonstrated that grass carp infection with A. hydrophila induced intestinal inflammation and impaired the structure and function of the intestinal mucosal barrier.
Collapse
Affiliation(s)
- Wei-Guang Kong
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovation Center, Wuhan, 430070, China
| | - Si-Si Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovation Center, Wuhan, 430070, China
| | - Xiao-Xuan Chen
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovation Center, Wuhan, 430070, China
| | - Yu-Qing Huang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovation Center, Wuhan, 430070, China
| | - Ying Tang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
- Freshwater Aquaculture Collaborative Innovation Center, Wuhan, 430070, China
| | - Zhi-Xin Wu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China.
- Freshwater Aquaculture Collaborative Innovation Center, Wuhan, 430070, China.
| |
Collapse
|
19
|
Soliman GA, Steenson SM, Etekpo AH. Effects of Metformin and a Mammalian Target of Rapamycin (mTOR) ATP-Competitive Inhibitor on Targeted Metabolomics in Pancreatic Cancer Cell Line. ACTA ACUST UNITED AC 2016; 6. [PMID: 28217402 DOI: 10.4172/2153-0769.1000183] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pancreatic Cancer (PC) is a devastating lethal disease. Therefore, there is an urgent need to develop new intervention strategies. The mammalian Target of Rapamycin (mTOR) is a conserved kinase and master regulator of metabolism and cell growth. mTOR is dysregulated in chronic diseases including diabetes and pancreatic cancer. Recent reports indicate that 50% of Pancreatic Ductal Adenocarcinoma (PDAC) patients are diabetic at the time of diagnosis. Furthermore, the anti-diabetic drug, metformin, which indirectly inhibits mTOR, has emerged as a potential therapeutic target for PC. The objective of this study is to determine the targeted-metabolomics profile in PDAC cell line (HPAF-II) with mTOR inhibition and the interaction between mTOR ATP-competitive inhibitor (Torin 2) and metformin as potential combined therapy in PC. HPAF-II cell lines were cultured in the presence of either Torin 2, metformin, both, or control vehicle. We utilized targeted LC/MS/MS to characterize the alterations in glycolytic and tricarboxylic acid cycle metabolomics, and employed Western Blot analysis for cell signaling activation by phosphorylation. Comparisons between groups were analyzed using one-way Analysis of Variance followed by secondary post-hoc analysis. After 1 h incubation with metformin, AMP concentration was significantly increased compared to other groups (p<0.03). After 24 h, Torin-2 significantly decreased glycolysis intermediates (fructose 1,6-bisphosphate (FBP), and 2-phosphoglycerate/3-phosphoglycerate), TCA intermediate metabolites (citrate/isocitrate, and malate), as well as Nicotinamide Adenine Dinucleotide (NAD+) and Flavin Adenine Dinucleotide (FAD), and ATP levels. When HPAF-II cells were incubated with both Torin-2 and metformin, there was a significant reduction in NAD+ and FAD, suggesting decreased levels of the energy equivalents that are available to the electron transport chain. Targeted metabolomics data indicate that mTOR complexes inhibition by Torin 2 reduced glycolytic intermediates and TCA metabolites in HPAF- II and may synergize with metformin to decrease the electron acceptors NAD+ and FAD which may lead to reduced energy production.
Collapse
Affiliation(s)
- Ghada A Soliman
- Department of Health Promotion, Social and Behavioral Health College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, 68198 USA
| | - Sharalyn M Steenson
- Department of Health Promotion, Social and Behavioral Health College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, 68198 USA
| | - Asserewou H Etekpo
- Department of Health Promotion, Social and Behavioral Health College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, 68198 USA
| |
Collapse
|
20
|
Sankapal S, Gupta P, Ratner D, Ding M, Shen C, Sanyal A, Stolz D, Cu-Uvin S, Ramratnam B, Chen Y. HIV Exposure to the Epithelia in Ectocervical and Colon Tissues Induces Inflammatory Cytokines Without Tight Junction Disruption. AIDS Res Hum Retroviruses 2016; 32:1054-1066. [PMID: 27153934 DOI: 10.1089/aid.2015.0185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Epithelial cells in human cervical and colonic mucosa do not express HIV receptor. However, HIV transmission occurs across the unbreached epithelia by an unknown mechanism. In this study, the effect of HIV exposure on tight junction (TJ) and cytokine production in ectocervical and colon mucosal epithelia in tissue biopsies was investigated in an organ culture model. After HIV exposure, the distribution patterns and quantities of epithelial TJ and adherens proteins were evaluated by immunofluorescence staining followed by confocal microscopy. Cytokine mRNA in the mucosal epithelia was also evaluated by real-time reverse transcription-polymerase chain reaction (RT-PCR). HIV transmission was evaluated by measuring p24 production in culture supernatant. Our results showed there were no significant changes in the distribution and quantities of epithelial TJ/adherens junction (AJ) proteins after exposure to HIV. However, higher levels of CXCL10 and CXCL11 mRNA expression were detected in HIV-exposed ectocervical epithelia. In case of colon mucosa, higher levels of CXCL10 and IL-6 mRNA expression were detected in HIV-exposed colon mucosa. Our study suggests that HIV induces cytokine production in epithelial cells, which may facilitate HIV transmission by recruiting HIV target cells in the submucosal region. Furthermore, HIV transmission may not occur through epithelial TJ/AJ disruption.
Collapse
Affiliation(s)
- Soni Sankapal
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Phalguni Gupta
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Deena Ratner
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ming Ding
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chengli Shen
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anwesha Sanyal
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Donna Stolz
- Department of Cell Biology and Physiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Susan Cu-Uvin
- Division of Infectious Diseases, Department of Medicine, Miriam Hospital/Alpert Medical School, Brown University, Providence, Rhode Island
- Division of Infectious Diseases, Department of Obstetrics and Gynecology, Miriam Hospital/Alpert Medical School, Brown University, Providence, Rhode Island
| | - Bharat Ramratnam
- Division of Infectious Diseases, Department of Medicine, Miriam Hospital/Alpert Medical School, Brown University, Providence, Rhode Island
- Division of Infectious Diseases, Department of Obstetrics and Gynecology, Miriam Hospital/Alpert Medical School, Brown University, Providence, Rhode Island
| | - Yue Chen
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
21
|
β1-Na(+),K(+)-ATPase gene therapy upregulates tight junctions to rescue lipopolysaccharide-induced acute lung injury. Gene Ther 2016; 23:489-99. [PMID: 26910760 DOI: 10.1038/gt.2016.19] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/08/2016] [Accepted: 01/19/2016] [Indexed: 01/05/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are associated with diverse disorders and characterized by disruption of the alveolar-capillary barrier, leakage of edema fluid into the lung, and substantial inflammation leading to acute respiratory failure. Gene therapy is a potentially powerful approach to treat ALI/ARDS through repair of alveolar epithelial function. Herein, we show that delivery of a plasmid expressing β1-subunit of the Na(+),K(+)-ATPase (β1-Na(+),K(+)-ATPase) alone or in combination with epithelial sodium channel (ENaC) α1-subunit using electroporation not only protected from subsequent lipopolysaccharide (LPS)-mediated lung injury, but also treated injured lungs. However, transfer of α1-subunit of ENaC (α1-ENaC) alone only provided protection benefit rather than treatment benefit although alveolar fluid clearance had been remarkably enhanced. Gene transfer of β1-Na(+),K(+)-ATPase, but not α1-ENaC, not only enhanced expression of tight junction protein zona occludins-1 (ZO-1) and occludin both in cultured cells and in mouse lungs, but also reduced pre-existing increase of lung permeability in vivo. These results demonstrate that gene transfer of β1-Na(+),K(+)-ATPase upregulates tight junction formation and therefore treats lungs with existing injury, whereas delivery of α1-ENaC only maintains pre-existing tight junction but not for generation. This indicates that the restoration of epithelial/endothelial barrier function may provide better treatment of ALI/ARDS.
Collapse
|
22
|
Lee JD, Yang WK, Lee MH. Impaired Na(+)/K(+)-ATPase Function in Patients with Interstitial Cystitis/Painful Bladder Syndrome. J Korean Med Sci 2016; 31:280-5. [PMID: 26839484 PMCID: PMC4729510 DOI: 10.3346/jkms.2016.31.2.280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/02/2015] [Indexed: 12/29/2022] Open
Abstract
Na(+)/K(+)-ATPase (NKA) is abundantly expressed in the basolateral membrane of epithelial cells, which is necessary for tight junction formation. The tight junction is an urothelial barrier between urine and the underlying bladder. Impairment of tight junctions allows migration of urinary solutes in patients with interstitial cystitis/painful bladder syndrome (IC/PBS). We evaluated NKA expression and activity in bladder samples from patients with IC/PBS. The study group consisted of 85 patients with IC/PBS, and the control group consisted of 20 volunteers. Bladder biopsies were taken from both groups. We determined the expression and distribution of NKA using NKA activity assays, immunoblotting, immunohistochemical staining, and immunofluorescent staining. The protein levels and activity of NKA in the study group were significantly lower than the control group (1.08 ± 0.06 vs. 2.39 ± 0.29 and 0.60 ± 0.04 vs. 1.81 ± 0.18 µmol ADP/mg protein/hour, respectively; P < 0.05). Additionally, immunofluorescent staining for detection of CK7, a marker of the bladder urothelium, predominantly colocalized with NKA in patients in the study group. Our results demonstrated the expression and activity of NKA were decreased in bladder biopsies of patients with IC/PBS. These findings suggest that NKA function is impaired in the bladders from patients with IC/PBS.
Collapse
Affiliation(s)
- Jane-Dar Lee
- Division of Urology, Department of Surgery, Taichung Armed Forces General Hospital, Taichung, Taiwan, Republic of China
- Central Taiwan University of Science and Technology, Taichung, Taiwan, Republic of China
- National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Wen-Kai Yang
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Ming-Huei Lee
- Department of Urology, Feng-Yuan Hospital, Ministry of Health and Welfare, Taichung, Taiwan, Republic of China
| |
Collapse
|
23
|
Balasubramaniam SL, Gopalakrishnapillai A, Barwe SP. Ion dependence of Na-K-ATPase-mediated epithelial cell adhesion and migration. Am J Physiol Cell Physiol 2015; 309:C437-41. [PMID: 26157008 DOI: 10.1152/ajpcell.00140.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Sona Lakshme Balasubramaniam
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware; and Department of Biological Sciences, University of Delaware, Newark, Delaware
| | | | - Sonali P Barwe
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware; and Department of Biological Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
24
|
Brune K, Frank J, Schwingshackl A, Finigan J, Sidhaye VK. Pulmonary epithelial barrier function: some new players and mechanisms. Am J Physiol Lung Cell Mol Physiol 2015; 308:L731-45. [PMID: 25637609 DOI: 10.1152/ajplung.00309.2014] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/27/2015] [Indexed: 12/20/2022] Open
Abstract
The pulmonary epithelium serves as a barrier to prevent access of the inspired luminal contents to the subepithelium. In addition, the epithelium dictates the initial responses of the lung to both infectious and noninfectious stimuli. One mechanism by which the epithelium does this is by coordinating transport of diffusible molecules across the epithelial barrier, both through the cell and between cells. In this review, we will discuss a few emerging paradigms of permeability changes through altered ion transport and paracellular regulation by which the epithelium gates its response to potentially detrimental luminal stimuli. This review is a summary of talks presented during a symposium in Experimental Biology geared toward novel and less recognized methods of epithelial barrier regulation. First, we will discuss mechanisms of dynamic regulation of cell-cell contacts in the context of repetitive exposure to inhaled infectious and noninfectious insults. In the second section, we will briefly discuss mechanisms of transcellular ion homeostasis specifically focused on the role of claudins and paracellular ion-channel regulation in chronic barrier dysfunction. In the next section, we will address transcellular ion transport and highlight the role of Trek-1 in epithelial responses to lung injury. In the final section, we will outline the role of epithelial growth receptor in barrier regulation in baseline, acute lung injury, and airway disease. We will then end with a summary of mechanisms of epithelial control as well as discuss emerging paradigms of the epithelium role in shifting between a structural element that maintains tight cell-cell adhesion to a cell that initiates and participates in immune responses.
Collapse
Affiliation(s)
- Kieran Brune
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
| | - James Frank
- The Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco VA Medical Center, and NCIRE/Veterans Health Research Institute, San Francisco, California
| | - Andreas Schwingshackl
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - James Finigan
- Division of Oncology, Cancer Center, National Jewish Health, Denver, Colorado
| | - Venkataramana K Sidhaye
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland;
| |
Collapse
|
25
|
Liang GH, Weber CR. Molecular aspects of tight junction barrier function. Curr Opin Pharmacol 2014; 19:84-9. [PMID: 25128899 DOI: 10.1016/j.coph.2014.07.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 12/01/2022]
Abstract
In complex multicellular organisms, epithelia lining body cavities regulate absorption and secretion of ions, organic molecules, and water. Proper function of epithelia depends on apically and basolaterally situated ion channels as well as tight junctions which seal the apical intercellular space. Without tight junctions, transepithelial concentration gradients of ions and nutrients would be dissipated through the paracellular space. Elevated tight junction permeability is a feature of many diseases of multiple organs, including the gastrointestinal tract [1,2,3(•),4(•)], kidney [5,6], and lungs [7,8]. In the intestines, epithelial barrier dysfunction is a major contributor to diarrhea and malnutrition and is associated with significant morbidity and mortality worldwide.
Collapse
Affiliation(s)
- Guo Hua Liang
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
26
|
Lubarski I, Asher C, Garty H. Modulation of cell polarization by the Na+-K+-ATPase-associated protein FXYD5 (dysadherin). Am J Physiol Cell Physiol 2014; 306:C1080-8. [PMID: 24717576 DOI: 10.1152/ajpcell.00042.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
FXYD5 (dysadherin or also called a related to ion channel, RIC) is a transmembrane auxiliary subunit of the Na(+)-K(+)-ATPase shown to increase its maximal velocity (Vmax). FXYD5 has also been identified as a cancer-associated protein whose expression in tumor-derived cell lines impairs cytoskeletal organization and increases cell motility. Previously, we have demonstrated that the expression of FXYD5 in M1 cells derived from mouse kidney collecting duct impairs the formation of tight and adherence junctions. The current study aimed to further explore effects of FXYD5 at a single cell level. It was found that in M1, as well as three other cell lines, FXYD5 inhibits transformation of adhered single cells from the initial radial shape to a flattened, elongated shape in the first stage of monolayer formation. This is also correlated to less ordered actin cables and fewer focal points. Structure-function analysis has demonstrated that the transmembrane domain of FXYD5, and not its unique extracellular segment, mediates the inhibition of change in cell shape. This domain has been shown before to be involved in the association of FXYD5 with the Na(+)-K(+)-ATPase, which leads to the increase in Vmax. Furthermore, specific transmembrane point mutations in FXYD5 that either increase or decrease its effect on cell elongation had a corresponding effect on the coimmunoprecipitation of FXYD5 with α Na(+)-K(+)-ATPase. These findings lend support to the possibility that FXYD5 affects cell polarization through its transmembrane domain interaction with the Na(+)-K(+)-ATPase. Yet interaction of FXYD5 with other proteins cannot be excluded.
Collapse
Affiliation(s)
- Irina Lubarski
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| | - Carol Asher
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| | - Haim Garty
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
27
|
Bellmann C, Schreivogel S, Günther R, Dabrowski S, Schümann M, Wolburg H, Blasig IE. Highly conserved cysteines are involved in the oligomerization of occludin-redox dependency of the second extracellular loop. Antioxid Redox Signal 2014; 20:855-67. [PMID: 23923978 PMCID: PMC3924802 DOI: 10.1089/ars.2013.5288] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED The tight junction (TJ) marker occludin is a 4-transmembrane domain (TMD) protein with unclear physiological and pathological functions, interacting with other TJ proteins. It oligomerizes and is redox sensitive. However, oligomerization sites and mechanisms are unknown. AIMS To identify hypoxia-sensitive binding sites, we investigated the consequences of amino-acid substitutions of highly conserved cysteines in human occludin, under normal and hypoxic incubations. RESULTS (i) The extracellular loop 2 (ECL2) showed homophilic trans- and cis-association between opposing cells and along the cell membrane, respectively, caused by a loop properly folded via an intraloop disulfide bridge between the shielded C216 and C237. Hypoxia and reductants prevented the associations. (ii) C82 in TMD1 directly cis-associated without disulfide formation. (iii) C76 in TMD1 and C148 in TMD2 limited the trans-interaction; C76 also limited occludin-related paracellular tightness and changed the strand morphology of claudin-1. (iv) The diminished binding strength found after substituting C82, C216, or C237 was accompanied by increased occludin mobility in the cell membrane. INNOVATION The data enable the first experimentally proven structural model of occludin and its homophilic interaction sites, in which the ECL2, via intraloop disulfide formation, has a central role in occludin's hypoxia-sensitive oligomerization and to regulate the structure of TJs. CONCLUSION Our findings support the new concept that occludin acts as a hypoxiasensor and contributes toward regulating the TJ assembly redox dependently. This is of pathogenic relevance for tissue barrier injury with reducing conditions. The ECL2 disulfide might be a model for four TMD proteins in TJs with two conserved cysteines in an ECL.
Collapse
|
28
|
Hu YJ, Wang YD, Tan FQ, Yang WX. Regulation of paracellular permeability: factors and mechanisms. Mol Biol Rep 2013; 40:6123-42. [PMID: 24062072 DOI: 10.1007/s11033-013-2724-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 09/14/2013] [Indexed: 12/20/2022]
Abstract
Epithelial permeability is composed of transcellular permeability and paracellular permeability. Paracellular permeability is controlled by tight junctions (TJs). Claudins and occludin are two major transmembrane proteins in TJs, which directly determine the paracellular permeability to different ions or large molecules. Intracellular signaling pathways including Rho/Rho-associated protein kinase, protein kinase Cs, and mitogen-activated protein kinase, modulate the TJ proteins to affect paracellular permeability in response for diverse stimuli. Cytokines, growth factors and hormones in organism can regulate the paracellular permeability via signaling pathway. The transcellular transporters such as Na-K-ATPase, Na(+)-coupled transporters and chloride channels, can interact with paracellular transport and regulate the TJs. In this review, we summarized the factors affecting paracellular permeability and new progressions of the related mechanism in recent studies, and pointed out further research areas.
Collapse
Affiliation(s)
- Yan-Jun Hu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, People's Republic of China
| | | | | | | |
Collapse
|
29
|
Mony S, Lee SJ, Harper JF, Barwe SP, Langhans SA. Regulation of Na,K-ATPase β1-subunit in TGF-β2-mediated epithelial-to-mesenchymal transition in human retinal pigmented epithelial cells. Exp Eye Res 2013; 115:113-22. [PMID: 23810808 DOI: 10.1016/j.exer.2013.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 04/15/2013] [Accepted: 06/07/2013] [Indexed: 12/14/2022]
Abstract
Proliferative vitreo retinopathy (PVR) is associated with extracellular matrix membrane (ECM) formation on the neural retina and disruption of the multilayered retinal architecture leading to distorted vision and blindness. During disease progression in PVR, retinal pigmented epithelial cells (RPE) lose cell-cell adhesion, undergo epithelial-to-mesenchymal transition (EMT), and deposit ECM leading to tissue fibrosis. The EMT process is mediated via exposure to vitreous cytokines and growth factors such as TGF-β2. Previous studies have shown that Na,K-ATPase is required for maintaining a normal polarized epithelial phenotype and that decreased Na,K-ATPase function and subunit levels are associated with TGF-β1-mediated EMT in kidney cells. In contrast to the basolateral localization of Na,K-ATPase in most epithelia, including kidney, Na,K-ATPase is found on the apical membrane in RPE cells. We now show that EMT is also associated with altered Na,K-ATPase expression in RPE cells. TGF-β2 treatment of ARPE-19 cells resulted in a time-dependent decrease in Na,K-ATPase β1 mRNA and protein levels while Na,K-ATPase α1 levels, Na,K-ATPase activity, and intracellular sodium levels remained largely unchanged. In TGF-β2-treated cells reduced Na,K-ATPase β1 mRNA inversely correlated with HIF-1α levels and analysis of the Na,K-ATPase β1 promoter revealed a putative hypoxia response element (HRE). HIF-1α bound to the Na,K-ATPase β1 promoter and inhibiting the activity of HIF-1α blocked the TGF-β2 mediated Na,K-ATPase β1 decrease suggesting that HIF-1α plays a potential role in Na,K-ATPase β1 regulation during EMT in RPE cells. Furthermore, knockdown of Na,K-ATPase β1 in ARPE-19 cells was associated with a change in cell morphology from epithelial to mesenchymal and induction of EMT markers such as α-smooth muscle actin and fibronectin, suggesting that loss of Na,K-ATPase β1 is a potential contributor to TGF-β2-mediated EMT in RPE cells.
Collapse
Affiliation(s)
- Sridevi Mony
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Rockland Center I, 1701 Rockland Road, Wilmington, DE 19803, USA.
| | | | | | | | | |
Collapse
|
30
|
Civan MM. DIDS and the Janus-faced Na⁺-K⁺-activated ATPase. Focus on "DIDS inhibits Na-K-ATPase activity in porcine nonpigmented ciliary epithelial cells by a Src family kinase-dependent mechanism". Am J Physiol Cell Physiol 2013; 305:C479-80. [PMID: 23636455 DOI: 10.1152/ajpcell.00114.2013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
31
|
Barwe SP, Skay A, McSpadden R, Huynh TP, Langhans SA, Inge LJ, Rajasekaran AK. Na,K-ATPase β-subunit cis homo-oligomerization is necessary for epithelial lumen formation in mammalian cells. J Cell Sci 2012; 125:5711-20. [PMID: 23077177 DOI: 10.1242/jcs.108795] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Na,K-ATPase is a hetero-oligomer of an α- and a β-subunit. The α-subunit (Na,K-α) possesses the catalytic function, whereas the β-subunit (Na,K-β) has cell-cell adhesion function and is localized to the apical junctional complex in polarized epithelial cells. Earlier, we identified two distinct conserved motifs on the Na,K-β(1) transmembrane domain that mediate protein-protein interactions: a glycine zipper motif involved in the cis homo-oligomerization of Na,K-β(1) and a heptad repeat motif that is involved in the hetero-oligomeric interaction with Na,K-α(1). We now provide evidence that knockdown of Na,K-β(1) prevents lumen formation and induces activation of extracellular regulated kinases 1 and 2 (ERK1/2) mediated by phosphatidylinositol 3-kinase in MDCK cells grown in three-dimensional collagen cultures. These cells sustained cell proliferation in an ERK1/2-dependent manner and did not show contact inhibition at high cell densities, as revealed by parental MDCK cells. This phenotype could be rescued by wild-type Na,K-β(1) or heptad repeat motif mutant of Na,K-β(1), but not by the glycine zipper motif mutant that abrogates Na,K-β(1) cis homo-oligomerization. These studies suggest that Na,K-β(1) cis homo-oligomerization rather than hetero-oligomerization with Na,K-α(1) is involved in epithelial lumen formation. The relevance of these findings to pre-neoplastic lumen filling in epithelial cancer is discussed.
Collapse
Affiliation(s)
- Sonali P Barwe
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Chang JT, Lowery LA, Sive H. Multiple roles for the Na,K-ATPase subunits, Atp1a1 and Fxyd1, during brain ventricle development. Dev Biol 2012; 368:312-22. [PMID: 22683378 DOI: 10.1016/j.ydbio.2012.05.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 05/29/2012] [Accepted: 05/29/2012] [Indexed: 01/26/2023]
Abstract
Formation of the vertebrate brain ventricles requires both production of cerebrospinal fluid (CSF), and its retention in the ventricles. The Na,K-ATPase is required for brain ventricle development, and we show here that this protein complex impacts three associated processes. The first requires both the alpha subunit (Atp1a1) and the regulatory subunit, Fxyd1, and leads to formation of a cohesive neuroepithelium, with continuous apical junctions. The second process leads to modulation of neuroepithelial permeability, and requires Atp1a1, which increases permeability with partial loss of function and decreases it with overexpression. In contrast, fxyd1 overexpression does not alter neuroepithelial permeability, suggesting that its activity is limited to neuroepithelium formation. RhoA regulates both neuroepithelium formation and permeability, downstream of the Na,K-ATPase. A third process, likely to be CSF production, is RhoA-independent, requiring Atp1a1, but not Fxyd1. Consistent with a role for Na,K-ATPase pump function, the inhibitor ouabain prevents neuroepithelium formation, while intracellular Na(+) increases after Atp1a1 and Fxyd1 loss of function. These data include the first reported role for Fxyd1 in the developing brain, and indicate that the Na,K-ATPase regulates three aspects of brain ventricle development essential for normal function: formation of a cohesive neuroepithelium, restriction of neuroepithelial permeability, and production of CSF.
Collapse
Affiliation(s)
- Jessica T Chang
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, United States
| | | | | |
Collapse
|
33
|
Xia XM, Li BK, Xing SM, Ruan HL. Emodin promoted pancreatic claudin-5 and occludin expression in experimental acute pancreatitis rats. World J Gastroenterol 2012; 18:2132-9. [PMID: 22563203 PMCID: PMC3342614 DOI: 10.3748/wjg.v18.i17.2132] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 03/03/2012] [Accepted: 03/09/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of emodin on pancreatic claudin-5 and occludin expression, and pancreatic paracellular permeability in acute pancreatitis (AP).
METHODS: Experimental pancreatitis was induced by retrograde injection of 5% sodium taurocholate into the biliopancreatic duct. Emodin was injected via the external jugular vein 0 or 6 h after induction of AP. Rats from sham operation and AP groups were injected with normal saline at the same time. Samples of pancreas were obtained 6 or 12 h after drug administration. Pancreatic morphology was examined with hematoxylin and eosin staining. Pancreatic edema was estimated by measuring tissue water content. Tumor necrosis factor (TNF)-α and interleukin (IL)-6 level were measured by enzyme-linked immunosorbent assay. Pancreatic paracellular permeability was assessed by tissue dye extravasation. Expression of pancreatic claudin-5 and occludin was examined by immunohistology, quantitative real-time reverse transcriptase polymerase chain reaction and western blotting.
RESULTS: Pancreatic TNF-α and IL-6 levels, wet/dry ratio, dye extravasation, and histological score were significantly elevated at 3, 6 and 12 h following sodium taurocholate infusion; treatment with emodin prevented these changes at all time points. Immunostaining of claudin-5 and occludin was detected in rat pancreas, which was distributed in pancreatic acinar cells, ductal cells and vascular endothelial cells, respectively. Sodium taurocholate infusion significantly decreased pancreatic claudin-5 and occludin mRNA and protein levels at 3, 6 and 12 h, and that could be promoted by intravenous administration of emodin at all time points.
CONCLUSION: These results demonstrate that emodin could promote pancreatic claudin-5 and occludin expression, and reduce pancreatic paracellular permeability.
Collapse
|
34
|
Alterations in intrinsic membrane properties and the axon initial segment in a mouse model of Angelman syndrome. J Neurosci 2012; 31:17637-48. [PMID: 22131424 DOI: 10.1523/jneurosci.4162-11.2011] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The axon initial segment (AIS) is the site of action potential initiation in neurons. Recent studies have demonstrated activity-dependent regulation of the AIS, including homeostatic changes in AIS length, membrane excitability, and the localization of voltage-gated Na(+) channels. The neurodevelopmental disorder Angelman syndrome (AS) is usually caused by the deletion of small portions of the maternal copy of chromosome 15, which includes the UBE3A gene. A mouse model of AS has been generated and these mice exhibit multiple neurological abnormalities similar to those observed in humans. We examined intrinsic properties of pyramidal neurons in hippocampal area CA1 from AS model mice and observed alterations in resting membrane potential, threshold potential, and action potential amplitude. The altered intrinsic properties in the AS mice were correlated with significant increases in the expression of the α1 subunit of Na/K-ATPase (α1-NaKA), the Na(+) channel NaV1.6, and the AIS anchoring protein ankyrin-G, as well as an increase in length of the AIS. These findings are the first evidence for pathology of intrinsic membrane properties and AIS-specific changes in AS, a neurodevelopmental disorder associated with autism.
Collapse
|
35
|
Abstract
Intercellular tight junctions (TJs) exhibit a complex molecular architecture involving the regulated cointeraction of cytoplasmic adaptor proteins (e.g., zonula occludens) and integral membrane linker proteins (e.g., occludin and claudins). They provide structural integrity to epithelial and endothelial tissues and create highly polarized barriers essential to homeostatic maintenance within vertebrate physiological systems, while their dysregulation is an established pathophysiological hallmark of many diseases (e.g., cancer, stroke, and inflammatory lung disease). The junctional complex itself is a highly dynamic signaling entity wherein participant proteins constantly undergo a blend of regulatory modifications in response to diverse physiological and pathological cues, ultimately diversifying the overall adhesive properties of the TJ. Occludin, a 65-kDa tetraspan integral membrane protein, contributes to TJ stabilization and optimal barrier function. This paper reviews our current knowledge of how tissue occludin is specifically modified at the posttranscriptional and posttranslational levels in diverse circumstances, with associated consequences for TJ dynamics and epithelial/endothelial homeostasis. Mechanistic concepts such as splice variance and alternate promoter usage, proteolysis, phosphorylation, dimerization, and ubiquitination are comprehensively examined, and possible avenues for future investigation highlighted.
Collapse
|
36
|
Lubarski I, Asher C, Garty H. FXYD5 (dysadherin) regulates the paracellular permeability in cultured kidney collecting duct cells. Am J Physiol Renal Physiol 2011; 301:F1270-80. [PMID: 21900457 DOI: 10.1152/ajprenal.00142.2011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
FXYD5 (dysadherin or RIC) is a member of the FXYD family of single-span transmembrane proteins associated with the Na(+)-K(+)-ATPase. Several studies have demonstrated enhanced expression of FXYD5 during metastasis and effects on cell adhesion and motility. The current study examines effects of FXYD5 on the paracellular permeability in the mouse kidney collecting duct cell line M1. Expressing FXYD5 in these cells leads to a large decrease in amiloride-insensitive transepithelial electrical resistance as well as increased permeability to 4-kDa dextran. Impairment of cell-cell contact was also demonstrated by staining cells for the tight and adherence junction markers zonula occludens-1 and β-catenin, respectively. This is further supported by large expansions of the interstitial spaces, visualized in electron microscope images. Expressing FXYD5 in M1 cells resulted in a decrease in N-glycosylation of β1 Na(+)-K(+)-ATPase, while silencing it in H1299 cells had an opposite effect. This may provide a mechanism for the above effects, since normal glycosylation of β1 plays an important role in cell-cell contact formation (Vagin O, Tokhtaeva E, Sachs G. J Biol Chem 281: 39573-39587, 2006).
Collapse
Affiliation(s)
- Irina Lubarski
- Dept. of Biological Chemistry, The Weizmann Institute of Science, Israel
| | | | | |
Collapse
|
37
|
Giannatselis H, Calder M, Watson AJ. Ouabain stimulates a Na+/K+-ATPase-mediated SFK-activated signalling pathway that regulates tight junction function in the mouse blastocyst. PLoS One 2011; 6:e23704. [PMID: 21901128 PMCID: PMC3162003 DOI: 10.1371/journal.pone.0023704] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 07/23/2011] [Indexed: 01/22/2023] Open
Abstract
The Na(+)/K(+)-ATPase plays a pivotal role during preimplantation development; it establishes a trans-epithelial ionic gradient that facilitates the formation of the fluid-filled blastocyst cavity, crucial for implantation and successful pregnancy. The Na(+)/K(+)-ATPase is also implicated in regulating tight junctions and cardiotonic steroid (CTS)-induced signal transduction via SRC. We investigated the expression of SRC family kinase (SFK) members, Src and Yes, during preimplantation development and determined whether SFK activity is required for blastocyst formation. Embryos were collected following super-ovulation of CD1 or MF1 female mice. RT-PCR was used to detect SFK mRNAs encoding Src and Yes throughout preimplantation development. SRC and YES protein were localized throughout preimplantation development. Treatment of mouse morulae with the SFK inhibitors PP2 and SU6656 for 18 hours resulted in a reversible blockade of progression to the blastocyst stage. Blastocysts treated with 10(-3) M ouabain for 2 or 10 minutes and immediately immunostained for phosphorylation at SRC tyr418 displayed reduced phosphorylation while in contrast blastocysts treated with 10(-4) M displayed increased tyr418 fluorescence. SFK inhibition increased and SFK activation reduced trophectoderm tight junction permeability in blastocysts. The results demonstrate that SFKs are expressed during preimplantation development and that SFK activity is required for blastocyst formation and is an important mediator of trophectoderm tight junction permeability.
Collapse
Affiliation(s)
- Holly Giannatselis
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Michele Calder
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Andrew J. Watson
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
38
|
Li Z, Zhang Z, Xie JX, Li X, Tian J, Cai T, Cui H, Ding H, Shapiro JI, Xie Z. Na/K-ATPase mimetic pNaKtide peptide inhibits the growth of human cancer cells. J Biol Chem 2011; 286:32394-403. [PMID: 21784855 DOI: 10.1074/jbc.m110.207597] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cells contain a large pool of nonpumping Na/K-ATPase that participates in signal transduction. Here, we show that the expression of α1 Na/K-ATPase is significantly reduced in human prostate carcinoma as well as in several human cancer cell lines. This down-regulation impairs the ability of Na/K-ATPase to regulate Src-related signaling processes. A supplement of pNaKtide, a peptide derived from α1 Na/K-ATPase, reduces the activities of Src and Src effectors. Consequently, these treatments stimulate apoptosis and inhibit growth in cultures of human cancer cells. Moreover, administration of pNaKtide inhibits angiogenesis and growth of tumor xenograft. Thus, the new findings demonstrate the in vivo effectiveness of pNaKtide and suggest that the defect in Na/K-ATPase-mediated signal transduction may be targeted for developing new anticancer therapeutics.
Collapse
Affiliation(s)
- Zhichuan Li
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio 43614, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yang Y, Dai M, Wilson TM, Omelchenko I, Klimek JE, Wilmarth PA, David LL, Nuttall AL, Gillespie PG, Shi X. Na+/K+-ATPase α1 identified as an abundant protein in the blood-labyrinth barrier that plays an essential role in the barrier integrity. PLoS One 2011; 6:e16547. [PMID: 21304972 PMCID: PMC3031570 DOI: 10.1371/journal.pone.0016547] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 12/21/2010] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The endothelial-blood/tissue barrier is critical for maintaining tissue homeostasis. The ear harbors a unique endothelial-blood/tissue barrier which we term "blood-labyrinth-barrier". This barrier is critical for maintaining inner ear homeostasis. Disruption of the blood-labyrinth-barrier is closely associated with a number of hearing disorders. Many proteins of the blood-brain-barrier and blood-retinal-barrier have been identified, leading to significant advances in understanding their tissue specific functions. In contrast, capillaries in the ear are small in volume and anatomically complex. This presents a challenge for protein analysis studies, which has resulted in limited knowledge of the molecular and functional components of the blood-labyrinth-barrier. In this study, we developed a novel method for isolation of the stria vascularis capillary from CBA/CaJ mouse cochlea and provided the first database of protein components in the blood-labyrinth barrier as well as evidence that the interaction of Na(+)/K(+)-ATPase α1 (ATP1A1) with protein kinase C eta (PKCη) and occludin is one of the mechanisms of loud sound-induced vascular permeability increase. METHODOLOGY/PRINCIPAL FINDINGS Using a mass-spectrometry, shotgun-proteomics approach combined with a novel "sandwich-dissociation" method, more than 600 proteins from isolated stria vascularis capillaries were identified from adult CBA/CaJ mouse cochlea. The ion transporter ATP1A1 was the most abundant protein in the blood-labyrinth barrier. Pharmacological inhibition of ATP1A1 activity resulted in hyperphosphorylation of tight junction proteins such as occludin which increased the blood-labyrinth-barrier permeability. PKCη directly interacted with ATP1A1 and was an essential mediator of ATP1A1-initiated occludin phosphorylation. Moreover, this identified signaling pathway was involved in the breakdown of the blood-labyrinth-barrier resulting from loud sound trauma. CONCLUSIONS/SIGNIFICANCE The results presented here provide a novel method for capillary isolation from the inner ear and the first database on protein components in the blood-labyrinth-barrier. Additionally, we found that ATP1A1 interaction with PKCη and occludin was involved in the integrity of the blood-labyrinth-barrier.
Collapse
Affiliation(s)
- Yue Yang
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Min Dai
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Teresa M. Wilson
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Irina Omelchenko
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - John E. Klimek
- Proteomic Shared Resources, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Phillip A. Wilmarth
- Proteomic Shared Resources, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Larry L. David
- Proteomic Shared Resources, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Alfred L. Nuttall
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Otolaryngology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Peter G. Gillespie
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Xiaorui Shi
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Otolaryngology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
- The Institute of Microcirculation, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
40
|
Tokhtaeva E, Sachs G, Vagin O. Diverse pathways for maturation of the Na,K-ATPase β1 and β2 subunits in the endoplasmic reticulum of Madin-Darby canine kidney cells. J Biol Chem 2010; 285:39289-302. [PMID: 20937802 PMCID: PMC2998159 DOI: 10.1074/jbc.m110.172858] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 10/05/2010] [Indexed: 01/05/2023] Open
Abstract
Proper folding of the Na,K-ATPase β subunits followed by assembly with the α subunits is necessary for their export from the endoplasmic reticulum (ER). Here we examine roles of the ER lectin chaperone, calnexin, and non-lectin chaperone, BiP, in folding and quality control of the β(1) and β(2) subunits in Madin-Darby canine kidney cells. Short term prevention of glycan-calnexin interactions by castanospermine slightly increases ER retention of β(1), suggesting minor involvement of calnexin in subunit folding. However, both prolonged incubation with castanospermine and removal of N-glycosylation sites do not affect the α(1)-assembly or trafficking of β(1) but increase the amount of the β(1)-bound BiP, showing that BiP can compensate for calnexin in assisting β(1) folding. In contrast to β(1), prevention of either N-glycosylation or glycan-calnexin interactions abolishes the α(1)-assembly and export of β(2) from the ER despite increased β(2)-BiP binding. Mutations in the α(1)-interacting regions of β(1) and β(2) subunits impair α(1) assembly but do not affect folding of the β subunits tested by their sensitivity to trypsin. At the same time, these mutations increase the amount of β-bound BiP but not of β-bound calnexin and increase ER retention of both β-isoforms. BiP, therefore, prevents the ER export of folded but α(1)-unassembled β subunits. These α(1)-unassembled β subunits are degraded faster than α(1)-bound β subunits, preventing ER overload. In conclusion, folding of the β(1) and β(2) subunits is assisted predominantly by BiP and calnexin, respectively. Folded β(1) and β(2) either assemble with α(1) or bind BiP. The α(1)-bound β subunits traffic to the Golgi, whereas BiP-bound β subunits are retained and degraded in the ER.
Collapse
Affiliation(s)
- Elmira Tokhtaeva
- From the Department of Physiology, School of Medicine, UCLA and Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California 90073
| | - George Sachs
- From the Department of Physiology, School of Medicine, UCLA and Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California 90073
| | - Olga Vagin
- From the Department of Physiology, School of Medicine, UCLA and Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California 90073
| |
Collapse
|
41
|
Translocation of Pseudomonas aeruginosa from the intestinal tract is mediated by the binding of ExoS to an Na,K-ATPase regulator, FXYD3. Infect Immun 2010; 78:4511-22. [PMID: 20805335 DOI: 10.1128/iai.00428-10] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The intestinal tract is considered the most important reservoir of Pseudomonas aeruginosa in intensive care units (ICUs). Gut colonization by P. aeruginosa underlies the development of invasive infections such as gut-derived sepsis. Intestinal colonization by P. aeruginosa is associated with higher ICU mortality rates. The translocation of endogenous P. aeruginosa from the colonized intestinal tract is an important pathogenic phenomenon. Here we identify bacterial and host proteins associated with bacterial penetration through the intestinal epithelial barrier. We first show by comparative genomic hybridization analysis that the exoS gene, encoding the type III effector protein, ExoS, was specifically detected in a clinical isolate that showed higher virulence in silkworms following midgut injection. We further show using a silkworm oral infection model that exoS is required both for virulence and for bacterial translocation from the midgut to the hemolymph. Using a bacterial two-hybrid screen, we show that the mammalian factor FXYD3, which colocalizes with and regulates the function of Na,K-ATPase, directly binds ExoS. A pulldown assay revealed that ExoS binds to the transmembrane domain of FXYD3, which also interacts with Na,K-ATPase. Na,K-ATPase controls the structure and barrier function of tight junctions in epithelial cells. Collectively, our results suggest that ExoS facilitates P. aeruginosa penetration through the intestinal epithelial barrier by binding to FXYD3 and thereby impairing the defense function of tight junctions against bacterial penetration.
Collapse
|
42
|
Rajasekaran SA, Huynh TP, Wolle DG, Espineda CE, Inge LJ, Skay A, Lassman C, Nicholas SB, Harper JF, Reeves AE, Ahmed MM, Leatherman JM, Mullin JM, Rajasekaran AK. Na,K-ATPase subunits as markers for epithelial-mesenchymal transition in cancer and fibrosis. Mol Cancer Ther 2010; 9:1515-24. [PMID: 20501797 DOI: 10.1158/1535-7163.mct-09-0832] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) is an important developmental process, participates in tissue repair, and occurs during pathologic processes of tumor invasiveness, metastasis, and tissue fibrosis. The molecular mechanisms leading to EMT are poorly understood. Although it is well documented that transforming growth factor (TGF)-beta plays a central role in the induction of EMT, the targets of TGF-beta signaling are poorly defined. We have shown earlier that Na,K-ATPase beta(1)-subunit levels are highly reduced in poorly differentiated kidney carcinoma cells in culture and in patients' tumor samples. In this study, we provide evidence that Na,K-ATPase is a new target of TGF-beta(1)-mediated EMT in renal epithelial cells, a model system used in studies of both cancer progression and fibrosis. We show that following treatment with TGF-beta(1), the surface expression of the beta(1)-subunit of Na,K-ATPase is reduced, before well-characterized EMT markers, and is associated with the acquisition of a mesenchymal phenotype. RNAi-mediated knockdown confirmed the specific involvement of the Na,K-ATPase beta(1)-subunit in the loss of the epithelial phenotype and exogenous overexpression of the Na,K-ATPase beta(1)-subunit attenuated TGF-beta(1)-mediated EMT. We further show that both Na,K-ATPase alpha- and beta-subunit levels are highly reduced in renal fibrotic tissues. These findings reveal for the first time that Na,K-ATPase is a target of TGF-beta(1)-mediated EMT and is associated with the progression of EMT in cancer and fibrosis.
Collapse
Affiliation(s)
- Sigrid A Rajasekaran
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tokhtaeva E, Munson K, Sachs G, Vagin O. N-glycan-dependent quality control of the Na,K-ATPase beta(2) subunit. Biochemistry 2010; 49:3116-28. [PMID: 20199105 DOI: 10.1021/bi100115a] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bulky hydrophilic N-glycans stabilize the proper tertiary structure of glycoproteins. In addition, N-glycans comprise the binding sites for the endoplasmic reticulum (ER)-resident lectins that assist correct folding of newly synthesized glycoproteins. To reveal the role of N-glycans in maturation of the Na,K-ATPase beta(2) subunit in the ER, the effects of preventing or modifying the beta(2) subunit N-glycosylation on trafficking of the subunit and its binding to the ER lectin chaperone, calnexin, were studied in MDCK cells. Preventing N-glycosylation abolishes binding of the beta(2) subunit to calnexin and results in the ER retention of the subunit. Furthermore, the fully N-glycosylated beta(2) subunit is retained in the ER when glycan-calnexin interactions are prevented by castanospermine, showing that N-glycan-mediated calnexin binding is required for correct subunit folding. Calnexin binding persists for several hours after translation is stopped with cycloheximide, suggesting that the beta(2) subunit undergoes repeated post-translational calnexin-assisted folding attempts. Homology modeling of the beta(2) subunit using the crystal structure of the alpha(1)-beta(1) Na,K-ATPase shows the presence of a relatively hydrophobic amino acid cluster proximal to N-glycosylation sites 2 and 7. Combined, but not separate, removal of sites 2 and 7 dramatically impairs calnexin binding and prevents the export of the beta(2) subunit from the ER. Similarly, hydrophilic substitution of two hydrophobic amino acids in this cluster disrupts both beta(2)-calnexin binding and trafficking of the subunit to the Golgi. Therefore, the hydrophobic residues in the proximity of N-glycans 2 and 7 are required for post-translational calnexin binding to these N-glycans in incompletely folded conformers, which, in turn, is necessary for maturation of the Na,K-ATPase beta(2) subunit.
Collapse
Affiliation(s)
- Elmira Tokhtaeva
- Department of Physiology, School of Medicine, UCLA, and Veterans Administration Greater Los Angeles Health Care System, VAGLAHS/West LA, Building 113, Room 324, 11301 Wilshire Boulevard, Los Angeles, California 90073, USA
| | | | | | | |
Collapse
|
44
|
|
45
|
Abstract
OBJECTIVES The human pancreatic duct cell line, HPAF, has been shown previously to secrete Cl(-) in response to Ca(2+)-mobilizing stimuli. Our aim was to assess the capacity of HPAF cells to transport and secrete HCO3(-). METHODS HPAF cells were grown as confluent monolayers on permeable supports. Short-circuit current was measured by voltage clamp. Intracellular pH (pHi) was measured by microfluorometry in cells loaded with 2',7'-bis(2-carboxyethyl)-5(6)-carboxyfluorescein (BCECF). RESULTS In HCO3(-)-free solutions, ATP-evoked changes in short-circuit current were inhibited by bumetanide, and the recovery of pHi from acid loading was abolished by 5-(N-ethyl-N-isopropyl)-amiloride (EIPA). In the presence of HCO3(-), ATP-evoked secretion was no longer inhibited by bumetanide, and there was a strong EIPA-insensitive recovery from acid loading, which was inhibited by 4,4'-diisothiocyanatodihydrostilbene-2,2'-disulfonate (H2DIDS). ATP, but not forskolin, stimulated HCO3(-) efflux from the cells. CONCLUSIONS In the absence of HCO3(-), ATP-evoked Cl(-) secretion is driven by a basolateral Na(+)-K(+)-2Cl(-) cotransporter, and pH(i) is regulated by apical and basolateral Na(+)/H(+) exchangers. In the presence of HCO3(-), ATP-evoked secretion is sustained in the absence of Na(+)-K(+)-2Cl(-) cotransporter activity and is probably driven by basolateral Na(+)-HCO3(-) cotransport.
Collapse
|
46
|
Krupinski T, Beitel GJ. Unexpected roles of the Na-K-ATPase and other ion transporters in cell junctions and tubulogenesis. Physiology (Bethesda) 2009; 24:192-201. [PMID: 19509129 DOI: 10.1152/physiol.00008.2009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Recent work shows that transport-independent as well as transport-dependent functions of ion transporters, and in particular the Na-K-ATPase, are required for formation and maintenance of several intercellular junctions. Furthermore, these junctional and other nonjunctional functions of ion transporters contribute to development of epithelial tubes. Here, we consider what has been learned about the roles of ion pumps in formation of junctions and epithelial tubes in mammals, zebrafish, Drosophila, and C. elegans. We propose that asymmetric association of the Na-K-ATPase with cell junctions early in metazoan evolution enabled vectorial transcellular ion transport and control of intraorganismal environment. Ion transport-independent functions of the Na-K-ATPase arose as junctional complexes evolved.
Collapse
Affiliation(s)
- Thomas Krupinski
- Department of Biochemistry, Northwestern University, Evanston, Illinois, USA
| | | |
Collapse
|
47
|
Ivanov AI, Samarin SN, Bachar M, Parkos CA, Nusrat A. Protein kinase C activation disrupts epithelial apical junctions via ROCK-II dependent stimulation of actomyosin contractility. BMC Cell Biol 2009; 10:36. [PMID: 19422706 PMCID: PMC2685374 DOI: 10.1186/1471-2121-10-36] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2008] [Accepted: 05/07/2009] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Disruption of epithelial cell-cell adhesions represents an early and important stage in tumor metastasis. This process can be modeled in vitro by exposing cells to chemical tumor promoters, phorbol esters and octylindolactam-V (OI-V), known to activate protein kinase C (PKC). However, molecular events mediating PKC-dependent disruption of epithelial cell-cell contact remain poorly understood. In the present study we investigate mechanisms by which PKC activation induces disassembly of tight junctions (TJs) and adherens junctions (AJs) in a model pancreatic epithelium. RESULTS Exposure of HPAF-II human pancreatic adenocarcinoma cell monolayers to either OI-V or 12-O-tetradecanoylphorbol-13-acetate caused rapid disruption and internalization of AJs and TJs. Activity of classical PKC isoenzymes was responsible for the loss of cell-cell contacts which was accompanied by cell rounding, phosphorylation and relocalization of the F-actin motor nonmuscle myosin (NM) II. The OI-V-induced disruption of AJs and TJs was prevented by either pharmacological inhibition of NM II with blebbistatin or by siRNA-mediated downregulation of NM IIA. Furthermore, AJ/TJ disassembly was attenuated by inhibition of Rho-associated kinase (ROCK) II, but was insensitive to blockage of MLCK, calmodulin, ERK1/2, caspases and RhoA GTPase. CONCLUSION Our data suggest that stimulation of PKC disrupts epithelial apical junctions via ROCK-II dependent activation of NM II, which increases contractility of perijunctional actin filaments. This mechanism is likely to be important for cancer cell dissociation and tumor metastasis.
Collapse
Affiliation(s)
- Andrei I Ivanov
- Department of Medicine, University of Rochester, Rochester, NY 14642, USA.
| | | | | | | | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Na,K-ATPase is an oligomeric protein composed of alpha subunits, beta subunits and FXYD proteins. The catalytic alpha subunit hydrolyzes ATP and transports the cations. Increasing experimental evidence suggest that beta subunits and FXYD proteins essentially contribute to the variable physiological needs of Na,K-ATPase function in different tissues. RECENT FINDINGS Beta subunits have a crucial role in the structural and functional maturation of Na,K-ATPase and modulate its transport properties. The chaperone function of the beta subunit is essential, for example, in the formation of tight junctions and cell polarity. Recent studies suggest that beta subunits also have inherent functions, which are independent of Na,K-ATPase activity and which may be involved in cell-cell adhesiveness and in suppression of cell motility. As for FXYD proteins, they modulate Na,K-ATPase activity in a tissue-specific way, in some cases in close cooperation with posttranslational modifications such as phosphorylation. SUMMARY A better understanding of the multiple functional roles of the accessory subunits of Na,K-ATPase is crucial to appraise their influence on physiological processes and their implication in pathophysiological states.
Collapse
|
49
|
Kawedia JD, Jiang M, Kulkarni A, Waechter HE, Matlin KS, Pauletti GM, Menon AG. The protein kinase A pathway contributes to Hg2+-induced alterations in phosphorylation and subcellular distribution of occludin associated with increased tight junction permeability of salivary epithelial cell monolayers. J Pharmacol Exp Ther 2008; 326:829-37. [PMID: 18550693 PMCID: PMC2677297 DOI: 10.1124/jpet.107.135798] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Hg(2+) is commonly used as an inhibitor of many aquaporins during measurements of transcellular water transport. To investigate whether it could also act on the paracellular water transport pathway, we asked whether addition of Hg(2+) affected transport of radiolabeled probes through tight junctions of a salivary epithelial cell monolayer. Inclusion of 1 mM Hg(2+) decreased transepithelial electrical resistance by 8-fold and augmented mannitol and raffinose flux by 13-fold, which translated into an estimated 44% increase in pore radius at the tight junction. These Hg(2+)-induced effects could be partially blocked by the protein kinase A (PKA) inhibitor N-[2-((p-bromocinnamyl) amino) ethyl]-5-isoquinolinesulfonamide, 2HCl (H89), suggesting that both-PKA dependent and PKA-independent mechanisms contribute to tight junction regulation. Western blot analyses showed a 2-fold decrease in tight junction-associated occludin after Hg(2+) treatment and the presence of a novel hyperphosphorylated form of occludin in the cytoplasmic fraction. These findings were corroborated by confocal imaging. The results from this study reveal a novel contribution of the PKA pathway in Hg(2+)-induced regulation of tight junction permeability in the salivary epithelial barrier. Therapeutically, this could be explored for pharmacological intervention in the treatment of dry mouth, Sjögren's syndrome, and possibly other disorders of fluid transport.
Collapse
Affiliation(s)
- Jitesh D Kawedia
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0524, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Inge LJ, Rajasekaran SA, Yoshimoto K, Mischel PS, McBride W, Landaw E, Rajasekaran AK. Evidence for a potential tumor suppressor role for the Na,K-ATPase beta1-subunit. Histol Histopathol 2008; 23:459-67. [PMID: 18228203 PMCID: PMC2779022 DOI: 10.14670/hh-23.459] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The Na,K-ATPase, consisting of two essential subunits (alpha, beta), plays a critical role in the regulation of ion homeostasis in mammalian cells. Recent studies indicate that reduced expression of the beta1 isoform (NaK-beta1) is commonly observed in carcinoma and is associated with events involved in cancer progression. In this study, we present evidence that repletion of NaK-beta1 in Moloney sarcoma virus-transformed Madin-Darby canine kidney cells (MSV-MDCK), a highly tumorigenic cell line, inhibits anchorage independent growth and suppresses tumor formation in immunocompromised mice. Additionally, using an in vitro cell-cell aggregation assay, we showed that cell aggregates of NaK-beta1 subunit expressing MSV-MDCK cells have reduced extracellular regulated kinase (ERK) 1/2 activity compared with parental MSV-MDCK cells. Finally, using immunohistochemistry and fully quantitative image analysis approaches, we showed that the levels of phosphorylated ERK 1/2 are inversely correlated to the NaK-beta1 levels in the tumors. These findings reveal for the first time that NaK-beta1 has a potential tumor-suppressor function in epithelial cells.
Collapse
Affiliation(s)
- L J Inge
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, Jonsson Comprehensive Cancer Center-David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | | | | | | | | | | |
Collapse
|