1
|
Shi G, Lu X, Wang G, Qian K, Peng X, Zhu Y, Wu J, Ke H, Chen L, Zheng Y, Yang T, Shi X, Huang P. Antimicrobial Effects of Kelisha Capsule on Escherichia coli Induced Diarrhea in Mice. Drug Des Devel Ther 2025; 19:2779-2794. [PMID: 40231195 PMCID: PMC11995921 DOI: 10.2147/dddt.s511158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Background Kelisha capsule (KLSC), listed in the Chinese Pharmacopoeia, has been employed for the treatment of infectious diarrhea. Nevertheless, the precise mechanism of KLSC remains to be elucidated. Aim of the Study This work was to investigate the antibacterial mode and therapeutic mechanism of KLSC towards E. coli infected diarrhea. Materials and Methods HPLC identified the primary chemical constituents of KLSC. A model of diarrhea was induced via E. coli injection. The impact of KLSC on E. coli-induced diarrhea was evaluated using a diarrhea score in Babl/c mice. The integrity of the intestinal barrier was assessed through staining and measuring levels of specific proteins. Furthermore, immunofluorescence staining was conducted to identify tight junction proteins in the small intestinal tissue. The full-length 16S rRNA was used to examine gut microbiota. Network pharmacology, molecular docking, and molecular dynamic simulation were used to investigate the impact of KLSC on diarrhea-related inflammation and quantify the expression levels of IL-6 and TNF-α in the blood and small intestine. The in vivo antibacterial activity and mode of action of KLSC were also investigated using IVIS imaging, transmission electron microscopy, scanning electron microscopy, and molecular dynamic simulation. Results KLSC exhibited a positive effect on E. coli infected diarrhea. The content of IL-6 and TNF-α in mice with KLSC was significantly reduced. Additionally, KLSC could restore the intestinal barrier function by reversing small intestine structure and up-regulating the expression of tight junction proteins. The KLSC significantly inhibit pathogenic bacteria and restore the gut microbiota diversity. IVIS Imaging System was visually observed significant antibacterial efficacy of KLSC in vivo. The KLSC disrupted the cell membrane system of E. coli through the interaction between bioactive molecule and bilayer. Conclusion KLSC normalized the gut barrier function, reshaped gut microbiota balance, suppressed the inflammatory pathways, and inhibited the bacterial activity, thereby relieving the diarrhea caused by E. coli.
Collapse
Affiliation(s)
- Guolin Shi
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People’s Republic of China
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Xiao Lu
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People’s Republic of China
| | - Kai Qian
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Xiaokang Peng
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Yangyang Zhu
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Jiaxu Wu
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Haiyong Ke
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Li Chen
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Yuanhang Zheng
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Tao Yang
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Xiaoying Shi
- Post-Doctoral Research Center, Zhejiang SUKEAN Pharmaceutical Co., Ltd, Hangzhou, 311228, People’s Republic of China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People’s Republic of China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People’s Republic of China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, People’s Republic of China
| |
Collapse
|
2
|
Adams L, Li X, Burchmore R, Goodwin RJA, Wall DM. Microbiome-derived metabolite effects on intestinal barrier integrity and immune cell response to infection. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001504. [PMID: 39392674 PMCID: PMC11469068 DOI: 10.1099/mic.0.001504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
The gut microbiota exerts a significant influence on human health and disease. While compositional changes in the gut microbiota in specific diseases can easily be determined, we lack a detailed mechanistic understanding of how these changes exert effects at the cellular level. However, the putative local and systemic effects on human physiology that are attributed to the gut microbiota are clearly being mediated through molecular communication. Here, we determined the effects of gut microbiome-derived metabolites l-tryptophan, butyrate, trimethylamine (TMA), 3-methyl-4-(trimethylammonio)butanoate (3,4-TMAB), 4-(trimethylammonio)pentanoate (4-TMAP), ursodeoxycholic acid (UDCA), glycocholic acid (GCA) and benzoate on the first line of defence in the gut. Using in vitro models of intestinal barrier integrity and studying the interaction of macrophages with pathogenic and non-pathogenic bacteria, we could ascertain the influence of these metabolites at the cellular level at physiologically relevant concentrations. Nearly all metabolites exerted positive effects on barrier function, but butyrate prevented a reduction in transepithelial resistance in the presence of the pathogen Escherichia coli, despite inducing increased apoptosis and exerting increased cytotoxicity. Induction of IL-8 was unaffected by all metabolites, but GCA stimulated increased intra-macrophage growth of E. coli and tumour necrosis-alpha (TNF-α) release. Butyrate, 3,4-TMAB and benzoate all increased TNF-α release independent of bacterial replication. These findings reiterate the complexity of understanding microbiome effects on host physiology and underline that microbiome metabolites are crucial mediators of barrier function and the innate response to infection. Understanding these metabolites at the cellular level will allow us to move towards a better mechanistic understanding of microbiome influence over host physiology, a crucial step in advancing microbiome research.
Collapse
Affiliation(s)
- Lauren Adams
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
| | - Xiang Li
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
| | - Richard Burchmore
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
| | - Richard J. A. Goodwin
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Daniel M. Wall
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
| |
Collapse
|
3
|
Pasquier N, Jaulin F, Peglion F. Inverted apicobasal polarity in health and disease. J Cell Sci 2024; 137:jcs261659. [PMID: 38465512 PMCID: PMC10984280 DOI: 10.1242/jcs.261659] [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] [Indexed: 03/12/2024] Open
Abstract
Apicobasal epithelial polarity controls the functional properties of most organs. Thus, there has been extensive research on the molecular intricacies governing the establishment and maintenance of cell polarity. Whereas loss of apicobasal polarity is a well-documented phenomenon associated with multiple diseases, less is known regarding another type of apicobasal polarity alteration - the inversion of polarity. In this Review, we provide a unifying definition of inverted polarity and discuss multiple scenarios in mammalian systems and human health and disease in which apical and basolateral membrane domains are interchanged. This includes mammalian embryo implantation, monogenic diseases and dissemination of cancer cell clusters. For each example, the functional consequences of polarity inversion are assessed, revealing shared outcomes, including modifications in immune surveillance, altered drug sensitivity and changes in adhesions to neighboring cells. Finally, we highlight the molecular alterations associated with inverted apicobasal polarity and provide a molecular framework to connect these changes with the core cell polarity machinery and to explain roles of polarity inversion in health and disease. Based on the current state of the field, failure to respond to extracellular matrix (ECM) cues, increased cellular contractility and membrane trafficking defects are likely to account for most cases of inverted apicobasal polarity.
Collapse
Affiliation(s)
- Nicolas Pasquier
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
- Cell Adhesion and Cancer lab, University of Turku, FI-20520 Turku, Finland
| | - Fanny Jaulin
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
| | - Florent Peglion
- Collective Invasion Team, Inserm U-1279, Gustave Roussy, Villejuif F-94805, France
| |
Collapse
|
4
|
Baral B, Kandpal M, Ray A, Jana A, Yadav DS, Sachin K, Mishra A, Baig MS, Jha HC. Helicobacter pylori and Epstein-Barr virus infection in cell polarity alterations. Folia Microbiol (Praha) 2024; 69:41-57. [PMID: 37672163 DOI: 10.1007/s12223-023-01091-7] [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: 02/19/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023]
Abstract
The asymmetrical distribution of the cellular organelles inside the cell is maintained by a group of cell polarity proteins. The maintenance of polarity is one of the vital host defense mechanisms against pathogens, and the loss of it contributes to infection facilitation and cancer progression. Studies have suggested that infection of viruses and bacteria alters cell polarity. Helicobacter pylori and Epstein-Barr virus are group I carcinogens involved in the progression of multiple clinical conditions besides gastric cancer (GC) and Burkitt's lymphoma, respectively. Moreover, the coinfection of both these pathogens contributes to a highly aggressive form of GC. H. pylori and EBV target the host cell polarity complexes for their pathogenesis. H. pylori-associated proteins like CagA, VacA OipA, and urease were shown to imbalance the cellular homeostasis by altering the cell polarity. Similarly, EBV-associated genes LMP1, LMP2A, LMP2B, EBNA3C, and EBNA1 also contribute to altered cell asymmetry. This review summarized all the possible mechanisms involved in cell polarity deformation in H. pylori and EBV-infected epithelial cells. We have also discussed deregulated molecular pathways like NF-κB, TGF-β/SMAD, and β-catenin in H. pylori, EBV, and their coinfection that further modulate PAR, SCRIB, or CRB polarity complexes in epithelial cells.
Collapse
Affiliation(s)
- Budhadev Baral
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Anushka Ray
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Ankit Jana
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Dhirendra Singh Yadav
- Central Forensic Science Laboratory, Pune, DFSS, Ministry of Home Affairs, Govt. of India, Talegaon MIDC Phase-1, Near JCB Factory, Pune, Maharashtra, 410506, India
| | - Kumar Sachin
- Himalayan School of Biosciences, Swami Rama Himalayan University, Swami Ram Nagar, Jolly Grant, Dehradun, Uttarakhand, 248 016, India
| | - Amit Mishra
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 65 Nagaur Road, Karwar, Jodhpur District, Rajasthan, 342037, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India.
| |
Collapse
|
5
|
Wang D, Jiang Q, Dong Z, Meng T, Hu F, Wang J, Yuan H. Nanocarriers transport across the gastrointestinal barriers: The contribution to oral bioavailability via blood circulation and lymphatic pathway. Adv Drug Deliv Rev 2023; 203:115130. [PMID: 37913890 DOI: 10.1016/j.addr.2023.115130] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
Oral administration is the preferred route of drug delivery in clinical practice due to its noninvasiveness, safety, convenience, and high patient compliance. The gastrointestinal tract (GIT) plays a crucial role in facilitating the targeted delivery of oral drugs. However, the GIT presents multiple barriers that impede drug absorption, including the gastric barrier in the stomach and the mucus and epithelial barriers in the intestine. In recent decades, nanotechnology has emerged as a promising approach for overcoming these challenges by utilizing nanocarrier-based drug delivery systems such as liposomes, micelles, polymeric nanoparticles, solid lipid nanoparticles, and inorganic nanoparticles. Encapsulating drugs within nanocarriers not only protects them from degradation but also enhances their transport and absorption across the GIT, ultimately improving oral bioavailability. The aim of this review is to elucidate the mechanisms underlying nanocarrier-mediated transportation across the GIT into systemic circulation via both the blood circulation and lymphatic pathway.
Collapse
Affiliation(s)
- Ding Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Qi Jiang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Zhefan Dong
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Jianwei Wang
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China; China Jinhua Institute of Zhejiang University, Jinhua 321299, PR China.
| |
Collapse
|
6
|
Van den Bossche S, Ostyn L, Vandendriessche V, Rigauts C, De Keersmaecker H, Nickerson CA, Crabbé A. The development and characterization of in vivo-like three-dimensional models of bronchial epithelial cell lines. Eur J Pharm Sci 2023; 190:106567. [PMID: 37633341 DOI: 10.1016/j.ejps.2023.106567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023]
Abstract
In vitro models of differentiated respiratory epithelium that allow high-throughput screening are an important tool to explore new therapeutics for chronic respiratory diseases. In the present study, we developed in vivo-like three-dimensional (3-D) models of bronchial epithelial cell lines that are commonly used to study chronic lung disease (16HBE14o-, CFBE41o- and CFBE41o- 6.2 WT-CFTR). To this end, cells were cultured on porous microcarrier beads in the rotating wall vessel (RWV) bioreactor, an optimized suspension culture method that allows higher throughput experimentation than other physiologically relevant models. Cell differentiation was compared to conventional two-dimensional (2-D) monolayer cultures and to the current gold standard in the respiratory field, i.e. air-liquid interface (ALI) cultures. Cellular differentiation was assessed in the three model systems by evaluating the expression and localization of markers that reflect the formation of tight junctions (zonula occludens 1), cell polarity (intercellular adhesion molecule 1 at the apical side and collagen IV expression at the basal cell side), multicellular complexity (acetylated α-tubulin for ciliated cells, CC10 for club cells, keratin-5 for basal cells) and mucus production (MUC5AC) through immunostaining and confocal laser scanning microscopy. Results were validated using Western Blot analysis. We found that tight junctions were expressed in 2-D monolayers, ALI cultures and 3-D models for all three cell lines. All tested bronchial epithelial cell lines showed polarization in ALI and 3-D cultures, but not in 2-D monolayers. Mucus secreting goblet-like cells were present in ALI and 3-D cultures of CFBE41o- and CFBE41o- 6.2 WT-CFTR cells, but not in 16HBE14o- cells. For all cell lines, there were no ciliated cells, basal cells, or club cells found in any of the model systems. In conclusion, we developed RWV-derived 3-D models of commonly used bronchial epithelial cell lines and showed that these models are a valuable alternative to ALI cultures, as they recapitulate similar key aspects of the in vivo parental tissue.
Collapse
Affiliation(s)
- Sara Van den Bossche
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Lisa Ostyn
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Valerie Vandendriessche
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Charlotte Rigauts
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Herlinde De Keersmaecker
- Centre of Advanced Light Microscopy, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium; Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium
| | - Cheryl A Nickerson
- School of Life Sciences, Biodesign Center for Fundamental and Applied Microbiomics, Biodesign Institute, Arizona State University, 727 E. Tyler Street, Tempe, Arizona 85281, USA
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Belgium.
| |
Collapse
|
7
|
Zhang K, Shen X, Han L, Wang M, Lian S, Wang K, Li C. Effects on the intestinal morphology, inflammatory response and microflora in piglets challenged with enterotoxigenic Escherichia coli K88. Res Vet Sci 2023; 157:50-61. [PMID: 36871456 DOI: 10.1016/j.rvsc.2023.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/17/2023] [Accepted: 02/25/2023] [Indexed: 03/03/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is an important cause of diarrhea in piglets, which leads to great economic losses. In this study, the ternary crossbred weaned piglets were orally administered with 1.5 × 1011 CFU ETEC K88 for three days. The results showed the ratio of villus length to crypt depth decreased in the duodenum and ileum after ETEC K88 infection. The expression of tight junction proteins ZO-1 in the jejunum and ileum, occludin in the jejunum and colon, and claudin-1 in the colon were down-regulated. The expression of IL-8 in the duodenum and jejunum, IL-13 in the colon, and TNF-α in the jejunum and colon were up-regulated. The expression of pBD1 in the colon, pBD2 in the jejunum, and pBD3 in the duodenum increased after infection. Meanwhile, the expression of TLR4, p38 MAPK and NF-κB p65 increased in all intestinal segments. Moreover, the expression of IL-8 in superficial cervical lymph nodes (SCLN), TNF-α in mesenteric lymph nodes (MLN), and IL-13 in inguinal lymph nodes (ILN) and MLN were up-regulated. The expression of pBD1 and pBD2 in SCLN and MLN, and pBD3 in SCLN were up-regulated. Acidobacteria and Proteobacteria were the most abundant phyla in both groups by analysis of intestinal microflora using 16 s rRNA sequencing, and the relative abundances of bacteria were found to be changed by Metastats software and LEfSe analysis. Our results indicated that cytokines and pBDs had different roles in different intestinal segments or different lymph nodes against ETEC K88, and gut microbiota was influenced after infection.
Collapse
Affiliation(s)
- Kun Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, Henan, People's Republic of China
| | - Xiaoyang Shen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, Henan, People's Republic of China
| | - Lu Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, Henan, People's Republic of China; Henan Animal Husbandry Service, Zhengzhou, Henan, People's Republic of China
| | - Mengyun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, Henan, People's Republic of China
| | - Shaoqiang Lian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, Henan, People's Republic of China
| | - Kejun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, Henan, People's Republic of China.
| | - Chunli Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, Henan, People's Republic of China.
| |
Collapse
|
8
|
Li X, Chen H, Zhang D. Discoidin domain receptor 1 may be involved in biological barrier homeostasis. J Clin Pharm Ther 2022; 47:2397-2407. [PMID: 35665520 DOI: 10.1111/jcpt.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 04/08/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase involved in the pathological processes of several diseases, such as keloid formation, renal fibrosis, atherosclerosis, tumours, and inflammatory processes. The biological barrier is the first line of defence against pathogens, and its disruption is closely related to diseases. In this review, we attempt to elucidate the relationship between DDR1 and the biological barrier, explore the potential biological value of DDR1, and review the current research status and clinical potential of DDR1-selective inhibitors. METHODS We conducted an extensive literature search on PubMed to collect studies on the relevance of DDR1 to biological barriers and DDR1-selective inhibitors. With these studies, we explored the relationship between DDR1 and biological barriers and briefly reviewed representative DDR1-selective inhibitors that have been reported in recent years. RESULTS AND DISCUSSION First, the review of the potential mechanisms by which DDR1 regulates biological barriers, including the epithelial, vascular, glomerular filtration, blood-labyrinth, and blood-brain barriers. In the body, DDR1 dysfunction and aberrant expression may be involved in the homeostasis of the biological barrier. Secondly, the review of DDR1 inhibitors reported in recent years shows that DDR1-targeted inhibition is an attractive and promising pharmacological intervention. WHAT IS NEW AND CONCLUSIONS This review shows that DDR1 is involved in various physiological and pathological processes and in the regulation of biological barrier homeostasis. However, studies on DDR1 and biological barriers are still scarce, and further studies are needed to elucidate their specific mechanisms. The development of targeted inhibitors provides a new direction and idea to study the mechanism of DDR1.
Collapse
Affiliation(s)
- Xiaoli Li
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Huiling Chen
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Department of Gastroenterology, Key Laboratory of Digestive Diseases, LanZhou University Second Hospital, LanZhou University, Lanzhou, China
| |
Collapse
|
9
|
Pickett MA, Sallee MD, Cote L, Naturale VF, Akpinaroglu D, Lee J, Shen K, Feldman JL. Separable mechanisms drive local and global polarity establishment in the Caenorhabditiselegans intestinal epithelium. Development 2022; 149:dev200325. [PMID: 36264257 PMCID: PMC9845746 DOI: 10.1242/dev.200325] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 10/06/2022] [Indexed: 11/17/2022]
Abstract
Apico-basolateral polarization is essential for epithelial cells to function as selective barriers and transporters, and to provide mechanical resilience to organs. Epithelial polarity is established locally, within individual cells to establish distinct apical, junctional and basolateral domains, and globally, within a tissue where cells coordinately orient their apico-basolateral axes. Using live imaging of endogenously tagged proteins and tissue-specific protein depletion in the Caenorhabditiselegans embryonic intestine, we found that local and global polarity establishment are temporally and genetically separable. Local polarity is initiated prior to global polarity and is robust to perturbation. PAR-3 is required for global polarization across the intestine but local polarity can arise in its absence, as small groups of cells eventually established polarized domains in PAR-3-depleted intestines in a HMR-1 (E-cadherin)-dependent manner. Despite the role of PAR-3 in localizing PKC-3 to the apical surface, we additionally found that PAR-3 and PKC-3/aPKC have distinct roles in the establishment and maintenance of local and global polarity. Taken together, our results indicate that different mechanisms are required for local and global polarity establishment in vivo.
Collapse
Affiliation(s)
- Melissa A. Pickett
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Department of Biological Sciences, San Jose State University, San Jose, CA 95112, USA
| | - Maria D. Sallee
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Lauren Cote
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | | | - Joo Lee
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Kang Shen
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
10
|
Meng HF, Jin J, Wang H, Wang LS, Wu CT. Recent advances in the therapeutic efficacy of hepatocyte growth factor gene-modified mesenchymal stem cells in multiple disease settings. J Cell Mol Med 2022; 26:4745-4755. [PMID: 35922965 PMCID: PMC9465188 DOI: 10.1111/jcmm.17497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/16/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022] Open
Abstract
Mesenchymal stem cell (MSC) therapy is considered a new treatment for a wide range of diseases and injuries, but challenges remain, such as poor survival, homing and engraftment rates, thus limiting the therapeutic efficacy of the transplanted MSCs. Many strategies have been developed to enhance the therapeutic efficacy of MSCs, such as preconditioning, co-transplantation with graft materials and gene modification. Hepatocyte growth factor (HGF) is secreted by MSCs, which plays an important role in MSC therapy. It has been reported that the modification of the HGF gene is beneficial to the therapeutic efficacy of MSCs, including diseases of the heart, lung, liver, urinary system, bone and skin, lower limb ischaemia and immune-related diseases. This review focused on studies involving HGF/MSCs both in vitro and in vivo. The characteristics of HGF/MSCs were summarized, and the mechanisms of their improved therapeutic efficacy were analysed. Furthermore, some insights are provided for HGF/MSCs' clinical application based on our understanding of the HGF gene and MSC therapy.
Collapse
Affiliation(s)
- Hong-Fang Meng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jide Jin
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hua Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Li-Sheng Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chu-Tse Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
11
|
Schulz E, Schumann M, Schneemann M, Dony V, Fromm A, Nagel O, Schulzke JD, Bücker R. Escherichia coli Alpha-Hemolysin HlyA Induces Host Cell Polarity Changes, Epithelial Barrier Dysfunction and Cell Detachment in Human Colon Carcinoma Caco-2 Cell Model via PTEN-Dependent Dysregulation of Cell Junctions. Toxins (Basel) 2021; 13:toxins13080520. [PMID: 34437391 PMCID: PMC8402498 DOI: 10.3390/toxins13080520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Escherichia coli (E. coli) of the B2 phylotype reside in human and animal intestines. The bacteria possess pathogenicity factors such as α-hemolysin (HlyA) that can induce intestinal epithelial leaks. We addressed the questions which host cell processes were dysregulated by E. coli HlyA that can potentiate intestinal diseases. The colon carcinoma cell line Caco-2 was infected by HlyA+ E. coli. Cell polarity regulation was analyzed by live cell imaging for the phosphatidylinositol-4,5-bisphosphate (PIP2) abundance. In Caco-2 monolayers, transepithelial electrical resistance was measured for characterization of barrier function. Cell proliferation and separation were assessed microscopically. Epithelial regulation and cell signaling were analyzed by RNA-Seq and Ingenuity Pathway Analysis (IPA). Our main findings from E. coli HlyA toxinogenicity in the colon carcinoma cell line are that (i) PIP2 at the membrane decrease, (ii) PTEN (phosphatase and tensin homolog) inhibition leads to cell polarity changes, (iii) epithelial leakiness follows these polarity changes by disruption of cell junctions and (iv) epithelial cell detachment increases. HlyA affected pathways, e.g., the PTEN and metastasis signaling, were identified by RNA-Seq bioinformatics calculations in IPA. In conclusion, HlyA affects cell polarity, thereby inducing epithelial barrier dysfunction due to defective tight junctions and focal leak induction as an exemplary mechanism for leaky gut.
Collapse
Affiliation(s)
- Emanuel Schulz
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (E.S.); (M.S.); (V.D.)
- Junior Clinician Scientist Program, Biomedical Innovation Academy, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Schumann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (E.S.); (M.S.); (V.D.)
| | - Martina Schneemann
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
| | - Violaine Dony
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (E.S.); (M.S.); (V.D.)
| | - Anja Fromm
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
| | - Oliver Nagel
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
| | - Jörg-Dieter Schulzke
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
| | - Roland Bücker
- Clinical Physiology/Nutritional Medicine, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (M.S.); (A.F.); (O.N.); (J.-D.S.)
- Correspondence: ; Tel.: +49-30-450-514548
| |
Collapse
|
12
|
Sebastián I, Okura N, Humbel BM, Xu J, Hermawan I, Matsuura C, Hall M, Takayama C, Yamashiro T, Nakamura S, Toma C. Disassembly of the apical junctional complex during the transmigration of Leptospira interrogans across polarized renal proximal tubule epithelial cells. Cell Microbiol 2021; 23:e13343. [PMID: 33864347 PMCID: PMC8459228 DOI: 10.1111/cmi.13343] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 12/15/2022]
Abstract
Bacterial pathogens have evolved multiple strategies to disassemble epithelial cell apical junctional complexes (AJCs) and infect epithelial cells. Leptospirosis is a widespread zoonotic infection, mainly caused by Leptospira interrogans, and its dissemination across host cell barriers is essential for its pathogenesis. However, the mechanism of bacterial dissemination across epithelial cell barriers remains poorly characterised. In this study, we analysed the interaction of L. interrogans with renal proximal tubule epithelial cells (RPTECs) and found that at 24 hr post‐infection, L. interrogans remain in close contact with the plasma membrane of the RPTEC by extracellularly adhering or crawling. Leptospira interrogans cleaved E‐cadherin and induced its endocytosis with release of the soluble N‐terminal fragment into the extracellular medium. Concomitantly, a gradual decrease in transepithelial electrical resistance (TEER), mislocalisation of AJC proteins (occludin, claudin‐10, ZO‐1, and cingulin) and cytoskeletal rearrangement were observed. Inhibition of clathrin‐mediated E‐cadherin endocytosis prevented the decrease in TEER. We showed that disassembly of AJCs in epithelial cells and transmigration of bacteria through the paracellular route are important for the dissemination of L. interrogans in the host.
Collapse
Affiliation(s)
- Isabel Sebastián
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Nobuhiko Okura
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Bruno M Humbel
- Imaging Section, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.,Microscopy Center, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jun Xu
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.,Department of Animal Microbiology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Idam Hermawan
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Chiaki Matsuura
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Malgorzata Hall
- Imaging Section, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Chitoshi Takayama
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Tetsu Yamashiro
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Shuichi Nakamura
- Department of Applied Physics, Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Claudia Toma
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
13
|
Choudhry N, Scott F, Edgar M, Sanger GJ, Kelly P. Reversal of Pathogen-Induced Barrier Defects in Intestinal Epithelial Cells by Contra-pathogenicity Agents. Dig Dis Sci 2021; 66:88-104. [PMID: 32034605 DOI: 10.1007/s10620-020-06121-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 01/29/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Environmental enteropathy (EE) is associated with stunting, impairment of responses to oral vaccines, and other adverse health consequences in young children throughout the developing world. EE is characterized by chronic low-grade intestinal inflammation and disrupted epithelial barrier integrity, partly resulting from dysregulation of tight junction proteins, observed in other enteropathies such as celiac disease. During EE, this dysregulation of tight junction expression amplifies translocation of pathogenic bacteria across the intestinal mucosa. AIMS The aim was to determine whether enteropathogen-mediated epithelial barrier failure can be ameliorated using contra-pathogenicity therapies. METHODS Intestinal epithelial barrier damage was assessed in Caco-2 cells incubated with three important enteropathogens identified in EE patients: Enteropathogenic Escherichia coli (EPEC), Citrobacter rodentium (C. rodentium), and Cryptosporidium parvum (C. parvum). Potential therapeutic molecules were tested to detect effects on transepithelial resistance (TER), bacterial translocation (BT), claudin-4 expression, and regulation of the inflammatory cytokine response. RESULTS All three enteropathogens compared to uninfected cells, reduced TER (EPEC; p < 0.0001, C. rodentium; p < 0.0001, C. parvum; p < 0.0007), reduced claudin-4 expression, and permitted BT (EPEC; p < 0.0001, C. rodentium; p < 0.0001, C. parvum; p < 0.0003) through the monolayer. Zinc, colostrum, epidermal growth factor, trefoil factor 3, resistin-like molecule-β, hydrocortisone, and the myosin light chain kinase inhibitor ML7 (Hexahydro-1-[(5-iodo-1-naphthalenyl)sulfonyl]-1H-1,4-diazepine hydrochloride); ML7) improved TER (up to 70%) and decreased BT (as much as 96%). Only zinc demonstrated modest antimicrobial activity. CONCLUSION The enteropathogens impaired intestinal-epithelial barrier integrity with dysregulation of claudin-4 and increased bacterial translocation. Enteropathogen-mediated damage was reduced using contra-pathogenicity agents which mitigated the effects of pathogens without direct antimicrobial activity.
Collapse
Affiliation(s)
- Naheed Choudhry
- Barts and The London School of Medicine, Queen Mary, University of London, London, E1 2AT, UK.
| | - Flora Scott
- Barts and The London School of Medicine, Queen Mary, University of London, London, E1 2AT, UK
| | - Meghan Edgar
- Gastroenterology Drug Discovery Unit, Takeda California, Inc., 10410 Science Center Drive, San Diego, CA, 92121, USA
| | - Gareth J Sanger
- Barts and The London School of Medicine, Queen Mary, University of London, London, E1 2AT, UK
- Gastroenterology Drug Discovery Unit, Takeda California, Inc., 10410 Science Center Drive, San Diego, CA, 92121, USA
| | - Paul Kelly
- Barts and The London School of Medicine, Queen Mary, University of London, London, E1 2AT, UK
| |
Collapse
|
14
|
Hua Y, Wu J, Fu M, Liu J, Li X, Zhang B, Zhao W, Wan C. Enterohemorrhagic Escherichia coli Effector Protein EspF Interacts With Host Protein ANXA6 and Triggers Myosin Light Chain Kinase (MLCK)-Dependent Tight Junction Dysregulation. Front Cell Dev Biol 2020; 8:613061. [PMID: 33425920 PMCID: PMC7785878 DOI: 10.3389/fcell.2020.613061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/02/2020] [Indexed: 01/06/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is an important foodborne pathogen that can cause bloody diarrhea and hemolytic uremic syndrome (HUS) in humans. EspF is one of the best-characterized effector proteins secreted from the type three secretion system to hijack host cell functions. However, the crucial pathogen-host interactions and the basis for the intestinal barrier disruption during infections remain elusive. Our previous study screened and verified the interaction between host protein ANXA6 and EspF protein. Here, by fluorescence resonance energy transfer (FRET) and co-immunoprecipitation (CO-IP), we verified that EspF interacts with ANXA6 through its C-terminal domain. Furthermore, we found that both the constitutive expression of EspF or ANXA6 and the co-expression of EspF-ANXA6 could decrease the levels of tight junction (TJ) proteins ZO-1 and occludin, and disrupt the distribution of ZO-1. Moreover, we showed that EspF-ANXA6 activated myosin light chain kinase (MLCK), induced the phosphorylation of myosin light chain (MLC) and PKCα, and down-regulated the expression level of Calmodulin protein. Collectively, this study revealed a novel interaction between the host protein (ANXA6) and EspF. The binding of EspF to ANXA6 may perturb TJs in an MLCK-MLC-dependent manner, and thus may be involved in EHEC pathogenic function.
Collapse
Affiliation(s)
- Ying Hua
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| | - Jiali Wu
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Muqing Fu
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jinyue Liu
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoxia Li
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Bao Zhang
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| | - Wei Zhao
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| | - Chengsong Wan
- Biosafety Level 3 Laboratory, Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, China.,Key Laboratory of Tropical Disease Research of Guangdong Province, Guangzhou, China
| |
Collapse
|
15
|
Hui W, Feng Y, Baoqi Y, Shuwei D, Ruihua X, Jiongjie H, Dongan C, Yan S, Shidong Z, Zuoting Y. Comparative proteomics analysis indicates that palmatine contributes to transepithelial migration by regulating cellular adhesion. PHARMACEUTICAL BIOLOGY 2020; 58:646-654. [PMID: 32658562 PMCID: PMC7470081 DOI: 10.1080/13880209.2020.1784961] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
CONTEXT Palmatine, a biologically active isoquinoline alkaloid, possesses multiple pharmaceutical activities against mucosal infection and inflammation. OBJECTIVE There are no reports about the influence of palmatine on uterine mucosal epithelial cells. MATERIALS AND METHODS We used proteomics to analyse differentially expressed proteins (DEPs) in goat endometrial epithelial cells (EECs) stimulated by lipopolysaccharide (LPS, 5 μg/mL, the dosage can induce inflammatory response, according to our previous study) for 12 h and then treated with palmatine (80 μg/mL) for 8 h; the dosage was selected based on MTT assay. The EECs without any treatment were used as controls. Every group was treated in triplicate. RESULTS A total of 428 DEPs in LPS-stimulated group and 486 DEPs in the palmatine-treated group were identified. Functional annotation analysis showed that palmatine mainly regulated the protein expression of structural molecules involved in the response to stimuli. Pathway analysis showed that cell adhesion molecule (CaM) pathways were most significant enriched due to palmatine treatment. Junction adhesion molecule 1 (JAM1), nectin 1 (NECT1) and cadherin 5 (CDH5), which play important roles in the transepithelial migration (TEpM) of leukocytes, were significantly downregulated by palmatine. Meanwhile, other proteins essential to the maintenance of cell adhesion and those that facilitate leukocyte migration were upregulated after palmatine treatment. Discussion and conclusions: The results suggested that palmatine regulates the expression of CaMs to affect TEpM during uterine mucosal inflammation and provides novel insight to understanding and developing palmatine pharmacology. Palmatine is a promising drug for treatment of mucosal inflammation.
Collapse
Affiliation(s)
- Wang Hui
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
| | - Yang Feng
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
| | - Yan Baoqi
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
| | - Dong Shuwei
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
| | - Xin Ruihua
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
| | - He Jiongjie
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
| | - Cui Dongan
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
| | - Sun Yan
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
- CONTACT Zhang Shidong
| | - Zhang Shidong
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
- CONTACT Zhang Shidong
| | - Yan Zuoting
- Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou, China
- Yan Zuoting Key Laboratory of New Animal Drug Project of Gansu Province/Key Laboratory of Veterinary Pharmaceutics Discovery, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Science, Lanzhou730050, China
| |
Collapse
|
16
|
Zheng M, Sun S, Zhou J, Liu M. Virulence factors impair epithelial junctions during bacterial infection. J Clin Lab Anal 2020; 35:e23627. [PMID: 33070380 PMCID: PMC7891540 DOI: 10.1002/jcla.23627] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/10/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial cells are typically connected through different types of cell junctions that are localized from the apical membrane to the basal surface. In this way, epithelium cells form the first barrier against pathogenic microorganisms and prevent their entry into internal organs and the circulatory system. Recent studies demonstrate that bacterial pathogens disrupt epithelial cell junctions through targeting junctional proteins by secreted virulence factors. In this review, we discuss the diverse strategies used by common bacterial pathogens, including Pseudomonas aeruginosa, Helicobacter pylori, and enteropathogenic Escherichia coli, to disrupt epithelial cell junctions during infection. We also discuss the potential of targeting the pathogenic mechanisms in the treatment of pathogen-associated diseases.
Collapse
Affiliation(s)
- Manxi Zheng
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Shuang Sun
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Min Liu
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
17
|
Bleomycin induced apical-basal polarity loss in alveolar epithelial cell contributes to experimental pulmonary fibrosis. Exp Cell Res 2020; 396:112295. [PMID: 32971116 DOI: 10.1016/j.yexcr.2020.112295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 08/26/2020] [Accepted: 09/16/2020] [Indexed: 11/23/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal fibrosing interstitial lung disease with limited therapeutic options and a median survival of 3 years after diagnosis. Dysregulated epithelial regeneration is key event involved in initiating and sustaining IPF. The type II alveolar epithelial cells (AECIIs) play a crucial role for epithelial regeneration and stabilisation of alveoli. Loss of cell apical-basal polarity contributes to fibrosis. AECII has apical-basal polarity, but it is poorly understood whether AECII apical-basal polarity loss is involved in fibrosis. Bleomycin is a traditional inducer of pulmonary fibrosis. Here firstly we observed that bleomycin induced apical-basal polarity loss in cultured AECIIs. Next, cell polarity proteins lethal (2) giant larvae 1 (Lgl1), PAR-3A, aPKC and PAR-6B were investigated. We found bleomycin induced increases of Lgl1 protein and decreases of PAR-3A protein, and bleomycin-induced PAR-3A depression was mediated by increased-Lgl1. Then Lgl1 siRNA was transfected into AECIIs. Lgl1 siRNA prevented apical-basal polarity loss in bleomycin-treated AECIIs. At last, Lgl1-conditional knockout mice were applied in making animal models. Bleomycin induced pulmonary fibrosis, but this was attenuated in Lgl1-conditional knockout mice. Together, these data indicated that bleomycin mediated AECII apical-basal polarity loss which contributed to experimental pulmonary fibrosis. Inhibition of Lgl1 should be a potential therapeutic strategy for the disease.
Collapse
|
18
|
Pickett MA, Naturale VF, Feldman JL. A Polarizing Issue: Diversity in the Mechanisms Underlying Apico-Basolateral Polarization In Vivo. Annu Rev Cell Dev Biol 2019; 35:285-308. [PMID: 31461314 DOI: 10.1146/annurev-cellbio-100818-125134] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Polarization along an apico-basolateral axis is a hallmark of epithelial cells and is essential for their selective barrier and transporter functions, as well as for their ability to provide mechanical resiliency to organs. Loss of polarity along this axis perturbs development and is associated with a wide number of diseases. We describe three steps involved in polarization: symmetry breaking, polarity establishment, and polarity maintenance. While the proteins involved in these processes are highly conserved among epithelial tissues and species, the execution of these steps varies widely and is context dependent. We review both theoretical principles underlying these steps and recent work demonstrating how apico-basolateral polarity is established in vivo in different tissues, highlighting how developmental and physiological contexts play major roles in the execution of the epithelial polarity program.
Collapse
Affiliation(s)
- Melissa A Pickett
- Department of Biology, Stanford University, Stanford, California 94305, USA;
| | - Victor F Naturale
- Department of Biology, Stanford University, Stanford, California 94305, USA;
| | - Jessica L Feldman
- Department of Biology, Stanford University, Stanford, California 94305, USA;
| |
Collapse
|
19
|
Abstract
The passive and regulated movement of ions, solutes, and water via spaces between cells of the epithelial monolayer plays a critical role in the normal intestinal functioning. This paracellular pathway displays a high level of structural and functional specialization, with the membrane-spanning complexes of the tight junctions, adherens junctions, and desmosomes ensuring its integrity. Tight junction proteins, like occludin, tricellulin, and the claudin family isoforms, play prominent roles as barriers to unrestricted paracellular transport. The past decade has witnessed major advances in our understanding of the architecture and function of epithelial tight junctions. While it has been long appreciated that microbes, notably bacterial and viral pathogens, target and disrupt junctional complexes and alter paracellular permeability, the precise mechanisms remain to be defined. Notably, renewed efforts will be required to interpret the available data on pathogen-mediated barrier disruption in the context of the most recent findings on tight junction structure and function. While much of the focus has been on pathogen-induced dysregulation of junctional complexes, commensal microbiota and their products may influence paracellular permeability and contribute to the normal physiology of the gut. Finally, microbes and their products have become important tools in exploring host systems, including the junctional properties of epithelial cells. © 2018 American Physiological Society. Compr Physiol 8:823-842, 2018.
Collapse
Affiliation(s)
- Jennifer Lising Roxas
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, USA
| | - V.K. Viswanathan
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, USA
- Department of Immunobiology, University of Arizona, Tucson, Arizona, USA
- BIO5 Institute for Collaborative Research, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|