1
|
Li X, Chen Y, Lan R, Liu P, Xiong K, Teng H, Tao L, Yu S, Han G. Transmembrane mucins in lung adenocarcinoma: understanding of current molecular mechanisms and clinical applications. Cell Death Discov 2025; 11:163. [PMID: 40210618 PMCID: PMC11985918 DOI: 10.1038/s41420-025-02455-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/13/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025] Open
Abstract
The mucin family is a group of highly glycosylated macromolecules widely present in human epithelial cells and with subtypes of secreted and membrane-associated forms. The membrane-associated mucins, known as transmembrane mucins, are not only involved in the formation of mucus barrier but also regulate cell signal transduction in physiological and pathological status. Transmembrane mucins could contribute to lung adenocarcinoma (LUAD) proliferation, apoptosis, angiogenesis, invasion, and metastasis, and remodel the immune microenvironment involved in immune escape. Furthermore, transmembrane mucins have been explored as potential LUAD indicators for diagnosis and prognosis. The development of targeted therapy and immunotherapeutic drugs targeting transmembrane mucins has also provided broad application prospects for clinic. In the following review, we summarize the characteristic structures of diverse transmembrane mucins, regulatory roles in promoting the progression of LUAD, and the current situation of diagnosis, prognosis, and therapeutic strategies based on transmembrane mucins.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Chen
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Lan
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Kai Xiong
- Department of Statistic, Inner Mongolia Forestry General Hospital, Yakeshi, China
| | - Hetai Teng
- Department of General Surgery, Inner Mongolia Forestry General Hospital, Yakeshi, China
| | - Lili Tao
- Department of Pathology, Peking University, Shenzhen Hospital, Shenzhen, China
| | - Shan Yu
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China.
- Heilongjiang Mental Hospital, Harbin, China.
| | - Guiping Han
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
2
|
Sahin AB, Karakurt S, Sezlev Bilecen D. Development of a mucoadhesive drug delivery system and its interaction with gastric cells. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:371-384. [PMID: 40099114 PMCID: PMC11912645 DOI: 10.3762/bjnano.16.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025]
Abstract
Drugs that are designed for local treatment of gastric diseases require increased gastric residence time for prolonged action and increased efficacy. In this study, we report a mucoadhesive drug delivery system that was developed to fulfill these requirements. Alginate nanoparticles were synthesized by water-in-oil emulsification followed by external gelation and then coated with the mucoadhesive polymer Eudragit RS100. The formulated nanoparticles had a mean size of 219 nm and positive charge. A peptide, as a model drug, was loaded onto the nanoparticles with an encapsulation efficiency of 58%. The release of the model drug from the delivery system was pH-independent and lasted for 7 days. The periodic acid-Schiff stain assay indicated 69% mucin interaction for the nanoparticles, which were also capable of diffusion through artificial mucus. The nanoparticles were not toxic to gastric epithelial cells and can be internalized by the cells within 4 h. The adsorption of nanoparticles onto mucus-secreting gastric cells was found to be correlated with cell number. The delivery system developed in this study is intended to be loaded with active therapeutic agents and has the potential to be used as an alternative drug delivery strategy for the treatment of gastric related diseases.
Collapse
Affiliation(s)
- Ahmet Baki Sahin
- Department of Molecular Biology and Genetics, Konya Food and Agriculture University, 42080, Konya, Türkiye
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, 34596, İstanbul, Türkiye
| | - Serdar Karakurt
- Department of Biochemistry, Faculty of Science, Selçuk University, 42130, Konya, Türkiye
| | - Deniz Sezlev Bilecen
- Department of Molecular Biology and Genetics, Konya Food and Agriculture University, 42080, Konya, Türkiye
| |
Collapse
|
3
|
Pisanu L, Mucaj K, Conio V, Bertuccio F, Giana I, Arlando L, Russo M, Montini S, Bortolotto C, Corsico AG, Stella GM. Lung bronchiectasisas a paradigm of the interplay between infection and colonization on plastic modulation of the pre-metastatic niche. Front Oncol 2024; 14:1480777. [PMID: 39469649 PMCID: PMC11513253 DOI: 10.3389/fonc.2024.1480777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
The lungs are most often a preferential target organ for malignant spreading and growth. It is well known that chronic parenchymal inflammation and prolonged injuries represents an independent risk factor for cancer onset. Growing evidence supports the implication of lung microbiota in the pathogenesis of lung cancer. However, the full interplay between chronic inflammation, bacterial colonization, pathologic condition as bronchiectasis and malignant growth deserves better clarification. We here aim at presenting and analyzing original data and discussing the state-of-the-art on the knowledge regarding how this complex milieu acts on the plasticity of the lung pre-metastatic niche to point out the rationale for early diagnosis and therapeutic targeting.
Collapse
Affiliation(s)
- Lucrezia Pisanu
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Klodjana Mucaj
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Valentina Conio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Francesco Bertuccio
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Ilaria Giana
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Lorenzo Arlando
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Marianna Russo
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Simone Montini
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Chandra Bortolotto
- Diagnostic Imaging and Radiotherapy Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia Medical School, Pavia, Italy
- Radiology Institute, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Angelo Guido Corsico
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Giulia Maria Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, Fondazione Istituto di Ricovero e Cura a carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
4
|
Lillehoj EP, Yu Y, Verceles AC, Imamura A, Ishida H, Piepenbrink KH, Goldblum SE. Stenotrophomonas maltophilia provokes NEU1-mediated release of a flagellin-binding decoy receptor that protects against lethal infection. iScience 2024; 27:110866. [PMID: 39314239 PMCID: PMC11418149 DOI: 10.1016/j.isci.2024.110866] [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: 04/10/2024] [Revised: 08/03/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Stenotrophomonas maltophilia (Sm), a multidrug-resistant pathogen often isolated from immunocompromised individuals, presents its flagellin to multimeric tandem repeats within the ectodomain of mucin-1 (MUC1-ED), expressed on airway epithelia. Flagellated Sm increases neuraminidase-1 (NEU1) sialidase association with and desialylation of MUC1-ED. This NEU1-mediated MUC1-ED desialylation unmasks cryptic binding sites for Sm flagellin, increasing flagellin and Sm binding to airway epithelia. MUC1 overexpression increases receptor number whereas NEU1 overexpression elevates receptor binding affinity. Silencing of either MUC1 or NEU1 reduces the flagellin-MUC1 interaction. Sm/flagellin provokes MUC1-ED autoproteolysis at a juxtamembranous glycine-serine peptide bond. MUC1-ED shedding from the epithelium not only occurs in vitro, but in the bronchoalveolar compartments of Sm/flagellin-challenged mice and patients with ventilator-associated Sm pneumonia. Finally, the soluble flagellin-targeting, MUC1-ED decoy receptor dose-dependently inhibits multiple Sm flagellin-driven pathogenic processes, in vitro, including motility, biofilm formation, adhesion, and proinflammatory cytokine production, and protects against lethal Sm lung infection, in vivo.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yafan Yu
- Department of Biochemistry, University of Nebraska, Lincoln, NE, USA
| | - Avelino C. Verceles
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Akihiro Imamura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Hideharu Ishida
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Kurt H. Piepenbrink
- Department of Biochemistry, University of Nebraska, Lincoln, NE, USA
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
- Department of Chemistry, University of Nebraska, Lincoln, NE, USA
| | - Simeon E. Goldblum
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Martins YA, Guerra-Gomes IC, Rodrigues TS, Tapparel C, Lopez RFV. Enhancing pulmonary delivery and immunomodulation of respiratory diseases through virus-mimicking nanoparticles. J Control Release 2024; 372:417-432. [PMID: 38908758 DOI: 10.1016/j.jconrel.2024.06.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
This study introduces the nanobromhexine lipid particle (NBL) platform designed for effective pulmonary drug delivery. Inspired by respiratory virus transport mechanisms, NBL address challenges associated with mucus permeation and inflammation in pulmonary diseases. Composed of low molecular weight polyethylene glycol-coated lipid nanoparticles with bromhexine hydrochloride, NBL exhibit a size of 118 ± 24 nm, a neutral zeta potential, osmolarity of 358 ± 28 mOsmol/kg, and a pH of 6.5. Nebulizing without leakage and showing no toxicity to epithelial cells, NBL display mucoadhesive properties with a 60% mucin-binding efficiency. They effectively traverse the dense mucus layer of Calu-3 cultures in an air-liquid interface, as supported by a 55% decrease in MUC5AC density and a 29% increase in nanoparticles internalization compared to non-exposed cells. In assessing immunomodulatory effects, NBL treatment in SARS-CoV-2-infected lung cells leads to a 40-fold increase in anti-inflammatory MUC1 gene expression, a proportional reduction in pro-inflammatory IL-6 expression, and elevated anti-inflammatory IL-10 expression. These findings suggest a potential mechanism to regulate the excessive IL-6 expression triggered by virus infection. Therefore, the NBL platform demonstrates promising potential for efficient pulmonary drug delivery and immunomodulation, offering a novel approach to addressing mucus permeation and inflammation in pulmonary diseases.
Collapse
Affiliation(s)
- Yugo Araújo Martins
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil
| | - Isabel Cristina Guerra-Gomes
- Fundação Oswaldo Cruz - FIOCRUZ, Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, SP 14040-030, Brazil
| | - Tamara Silva Rodrigues
- Department of Biochemistry and Imumunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Renata Fonseca Vianna Lopez
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil.
| |
Collapse
|
6
|
Choi M, Tichenor AA. Regional Conjunctival Differences in Glycocalyx Mucin Expression in Dry Eye and Normal Subjects. Invest Ophthalmol Vis Sci 2024; 65:20. [PMID: 38334701 PMCID: PMC10860684 DOI: 10.1167/iovs.65.2.20] [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: 07/12/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Purpose To compare regional conjunctival expression of membrane-associated mucins (MAMs) MUC1, MUC4, and MUC16 in normal and dry eye (DE) subjects. Methods Adults with and without signs and symptoms of DE were recruited. Impression cytology was performed to collect MAMs from four bulbar and upper eyelid palpebral conjunctival regions of both eyes. After protein extraction, samples from both eyes of a single subject were pooled by region, and expression was analyzed using a capillary electrophoresis nano-immunoassay system. The chemiluminescence intensity of each antigen binding signal was calculated after normalization to the total protein amount. Statistical analyses were conducted using GraphPad Prime 9. Results Samples from thirteen to sixteen DE and seven to eleven normal subjects were analyzed. In normal samples, MUC1 expression from the nasal bulbar conjunctiva was significantly greater than superior (P = 0.004) and inferior (P = 0.005). In DE samples, MUC1 expression was highest superiorly. Significant differences in MUC4 and MUC16 expression were not seen in normal samples. MUC4 and MUC16 expression was upregulated superiorly (P < 0.0001) and inferiorly (P < 0.0001) in DE compared with those regions in normal samples. Conclusions Although MAMs form a hydrophilic barrier called the glycocalyx, each mucin may have unique functions that are currently unexplored. All MAMs were expressed in the upper palpebral conjunctiva. Increased MUC1 expression nasally in healthy subjects suggests a functional need for increased protection. When comparing DE with normal eyes, upregulation of MUC1 superiorly, and in both MUC4 and MUC16 both superiorly and inferiorly, may indicate a need to decrease eyelid friction during blinking, especially in DE.
Collapse
Affiliation(s)
- Moonjung Choi
- New England College of Optometry, Boston, Massachusetts, United States
| | | |
Collapse
|
7
|
Lee S, Yang HK, Lee HJ, Park DJ, Kong SH, Park SK. Cross-phenotype association analysis of gastric cancer: in-silico functional annotation based on the disease-gene network. Gastric Cancer 2023; 26:517-527. [PMID: 36995485 DOI: 10.1007/s10120-023-01380-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/02/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND A gene or variant has pleiotropic effects, and genetic variant identification across multiple phenotypes can provide a comprehensive understanding of biological pathways shared among different diseases or phenotypes. Discovery of genetic loci associated with multiple diseases can simultaneously support general interventions. Several meta-analyses have shown genetic associations with gastric cancer (GC); however, no study has identified associations with other phenotypes using this approach. METHODS Here, we applied disease network analysis and gene-based analysis (GBA) to examine genetic variants linked to GC and simultaneously associated with other phenotypes. We conducted a single-nucleotide polymorphism (SNP) level meta-analysis and GBA through a systematic genome-wide association study (GWAS) linked to GC, to integrate published results for the SNP variants and group them into major GC-associated genes. We then performed disease network and expression quantitative trait loci (eQTL) analyses to evaluate cross-phenotype associations and expression levels of GC-related genes. RESULTS Seven genes (MTX1, GBAP1, MUC1, TRIM46, THBS3, PSCA, and ABO) were associated with GC as well as blood urea nitrogen (BUN), glomerular filtration rate (GFR), and uric acid (UA). In addition, 17 SNPs regulated the expression of genes located on 1q22, 24 SNPs regulated the expression of PSCA on 8q24.3, and rs7849820 regulated the expression of ABO on 9q34.2. Furthermore, rs1057941 and rs2294008 had the highest posterior causal probabilities of being a causal candidate SNP in 1q22, and 8q24.3, respectively. CONCLUSIONS These findings identified seven GC-associated genes exhibiting a cross-association with GFR, BUN, and UA.
Collapse
Affiliation(s)
- Sangjun Lee
- Department of Preventive Medicine, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Han-Kwang Yang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk-Joon Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Do Joong Park
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sue K Park
- Department of Preventive Medicine, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Korea.
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
8
|
Amalia R, Panenggak NSR, Doohan D, Rezkitha YAA, Waskito LA, Syam AF, Lubis M, Yamaoka Y, Miftahussurur M. A comprehensive evaluation of an animal model for Helicobacter pylori-associated stomach cancer: Fact and controversy. Helicobacter 2023; 28:e12943. [PMID: 36627714 DOI: 10.1111/hel.12943] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 01/12/2023]
Abstract
Even though Helicobacter pylori infection was the most causative factor of gastric cancer, numerous in vivo studies failed to induce gastric cancer using H. pylori infection only. The utilization of established animal studies in cancer research is crucial as they aim to investigate the coincidental association between suspected oncogenes and pathogenesis as well as generate models for the development and testing of potential treatments. The methods to establish gastric cancer using infected animal models remain limited, diverse in methods, and showed different results. This study investigates the differences in animal models, which highlight different pathological results in gaster by literature research. Electronic databases searched were performed in PubMed, Science Direct, and Cochrane, without a period filter. A total of 135 articles were used in this study after a full-text assessment was conducted. The most frequent animal models used for gastric cancer were Mice, while Mongolian gerbils and Transgenic mice were the most susceptible model for gastric cancer associated with H. pylori infection. Additionally, transgenic mice showed that the susceptibility to gastric cancer progression was due to genetic and epigenetic factors. These studies showed that in Mongolian gerbil models, H. pylori could function as a single agent to trigger stomach cancer. However, most gastric cancer susceptibilities were not solely relying on H. pylori infection, and numerous factors are involved in cancer progression. Further study using Mongolian gerbils and Transgenic mice is crucial to conduct and establish the best models for gastric cancer associated H. pylori.
Collapse
Affiliation(s)
- Rizki Amalia
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Nur Syahadati Retno Panenggak
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Dalla Doohan
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia.,Department of Anatomy, Histology and Pharmacology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Yudith Annisa Ayu Rezkitha
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia.,Department of Internal Medicine, Faculty of Medicine, Universitas Muhammadiyah Surabaya, Surabaya, Indonesia
| | - Langgeng Agung Waskito
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia.,Department of Physiology and Medical Biochemistry, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ari Fahrial Syam
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Masrul Lubis
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan.,Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Texas, Houston, USA
| | - Muhammad Miftahussurur
- Helicobacter pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia.,Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine-Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
9
|
Oruc A, Simsek G. A Pathophysiological Approach To Current Biomarkers. Biomark Med 2022. [DOI: 10.2174/9789815040463122010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Biomarkers are necessary for screening and diagnosing numerous diseases,
predicting the prognosis of patients, and following-up treatment and the course of the
patient. Everyday new biomarkers are being used in clinics for these purposes. This
section will discuss the physiological roles of the various current biomarkers in a
healthy person and the pathophysiological mechanisms underlying the release of these
biomarkers. This chapter aims to gain a new perspective for evaluating and interpreting
the most current biomarkers.
Collapse
Affiliation(s)
- Aykut Oruc
- Department of Physiology,Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpaşa,
Istanbul, Turkey
| | - Gonul Simsek
- Department of Physiology,Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpaşa,
Istanbul, Turkey
| |
Collapse
|
10
|
Lee S, Yang HK, Lee HJ, Park DJ, Kong SH, Park SK. Systematic review of gastric cancer-associated genetic variants, gene-based meta-analysis, and gene-level functional analysis to identify candidate genes for drug development. Front Genet 2022; 13:928783. [PMID: 36081994 PMCID: PMC9446437 DOI: 10.3389/fgene.2022.928783] [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: 04/26/2022] [Accepted: 07/25/2022] [Indexed: 12/05/2022] Open
Abstract
Objective: Despite being a powerful tool to identify novel variants, genome-wide association studies (GWAS) are not sufficient to explain the biological function of variants. In this study, we aimed to elucidate at the gene level the biological mechanisms involved in gastric cancer (GC) development and to identify candidate drug target genes. Materials and methods: We conducted a systematic review for GWAS on GC following the PRISMA guidelines. Single nucleotide polymorphism (SNP)-level meta-analysis and gene-based analysis (GBA) were performed to identify SNPs and genes significantly associated with GC. Expression quantitative trait loci (eQTL), disease network, pathway enrichment, gene ontology, gene-drug, and chemical interaction analyses were conducted to elucidate the function of the genes identified by GBA. Results: A review of GWAS on GC identified 226 SNPs located in 91 genes. In the comprehensive GBA, 44 genes associated with GC were identified, among which 12 genes (THBS3, GBAP1, KRTCAP2, TRIM46, HCN3, MUC1, DAP3, EFNA1, MTX1, PRKAA1, PSCA, and ABO) were eQTL. Using disease network and pathway analyses, we identified that PRKAA, THBS3, and EFNA1 were significantly associated with the PI3K-Alt-mTOR-signaling pathway, which is involved in various oncogenic processes, and that MUC1 acts as a regulator in both the PI3K-Alt-mTOR and P53 signaling pathways. Furthermore, RPKAA1 had the highest number of interactions with drugs and chemicals. Conclusion: Our study suggests that PRKAA1, a gene in the PI3K-Alt-mTOR-signaling pathway, could be a potential target gene for drug development associated with GC in the future. Systematic Review Registration: website, identifier registration number.
Collapse
Affiliation(s)
- Sangjun Lee
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, South Korea
| | - Han-Kwang Yang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyuk-Joon Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Do Joong Park
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Seong-Ho Kong
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Sue K. Park
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
11
|
Lillehoj EP, Luzina IG, Atamas SP. Mammalian Neuraminidases in Immune-Mediated Diseases: Mucins and Beyond. Front Immunol 2022; 13:883079. [PMID: 35479093 PMCID: PMC9035539 DOI: 10.3389/fimmu.2022.883079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 12/28/2022] Open
Abstract
Mammalian neuraminidases (NEUs), also known as sialidases, are enzymes that cleave off the terminal neuraminic, or sialic, acid resides from the carbohydrate moieties of glycolipids and glycoproteins. A rapidly growing body of literature indicates that in addition to their metabolic functions, NEUs also regulate the activity of their glycoprotein targets. The simple post-translational modification of NEU protein targets-removal of the highly electronegative sialic acid-affects protein folding, alters protein interactions with their ligands, and exposes or covers proteolytic sites. Through such effects, NEUs regulate the downstream processes in which their glycoprotein targets participate. A major target of desialylation by NEUs are mucins (MUCs), and such post-translational modification contributes to regulation of disease processes. In this review, we focus on the regulatory roles of NEU-modified MUCs as coordinators of disease pathogenesis in fibrotic, inflammatory, infectious, and autoimmune diseases. Special attention is placed on the most abundant and best studied NEU1, and its recently discovered important target, mucin-1 (MUC1). The role of the NEU1 - MUC1 axis in disease pathogenesis is discussed, along with regulatory contributions from other MUCs and other pathophysiologically important NEU targets.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Irina G. Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Research Service, Baltimore Veterans Affairs (VA) Medical Center, Baltimore, MD, United States
| | - Sergei P. Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Choudhary I, Vo T, Paudel K, Wen X, Gupta R, Kesimer M, Patial S, Saini Y. Vesicular and extravesicular protein analyses from the airspaces of ozone-exposed mice revealed signatures associated with mucoinflammatory lung disease. Sci Rep 2021; 11:23203. [PMID: 34853335 PMCID: PMC8636509 DOI: 10.1038/s41598-021-02256-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 11/08/2021] [Indexed: 12/16/2022] Open
Abstract
Lung epithelial lining fluid (ELF) harbors a variety of proteins that influence homeostatic and stress responses in the airspaces. Exosomes, nano-sized extracellular vesicles, contain many proteins that vary in abundance and composition based on the prevailing conditions. Ozone causes inflammatory responses in the airspaces of experimental animals and humans. However, the exosomal protein signatures contained within the ELF from ozone-exposed lung airspaces remain poorly characterized. To explore this, we hypothesized that ozone triggers the release of exosome-bound inflammatory proteins from various cells that reflect mucoobstructive lung disease. Accordingly, we repetitively exposed adult male and female C57BL/6 mice to HEPA-filtered air (air) or 0.8 ppm ozone (4 h per day) for 14 days (five consecutive days of exposure, 2 days of rest, five consecutive days of exposure, 2 days of rest, four consecutive days of exposure). Exosome-bound proteomic signatures, as well as the levels of soluble inflammatory mediators in the bronchoalveolar lavage fluid (BALF), were determined 12-16 h after the last exposure. Principal component analyses of the exosome-bound proteome revealed a clear distinction between air-exposed and ozone-exposed mice, as well as between ozone-exposed males and ozone-exposed females. In addition to 575 proteins that were enriched in both sexes upon ozone exposure, 243 and 326 proteins were enriched uniquely in ozone-exposed males and females, respectively. Ingenuity pathway analyses on enriched proteins between ozone- and air-exposed mice revealed enrichment of pro-inflammatory pathways. More specifically, macrophage activation-related proteins were enriched in exosomes from ozone-exposed mice. Cytokine analyses on the BALF revealed elevated levels of G-CSF, KC, IP-10, IL-6, and IL-5 in ozone-exposed mice. Finally, the histopathological assessment revealed significantly enhanced intracellular localization of mucoinflammatory proteins including MUC5B and FIZZ1 in ozone-exposed mice in a cell-specific manner indicating the cellular sources of the proteins that are ferried in the exosomes upon ozone-induced lung injury. Collectively, this study identified exosomal, secretory, and cell-specific proteins and biological pathways following repetitive exposure of mice to ozone.
Collapse
Affiliation(s)
- Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Thao Vo
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Kshitiz Paudel
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Xue Wen
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Richa Gupta
- Department of Pathology and Laboratory Medicine, UNC School of Medicine, Chapel Hill, NC, 27510, USA
| | - Mehmet Kesimer
- Department of Pathology and Laboratory Medicine, UNC School of Medicine, Chapel Hill, NC, 27510, USA
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
13
|
Martinez-Carrasco R, Argüeso P, Fini ME. Membrane-associated mucins of the human ocular surface in health and disease. Ocul Surf 2021; 21:313-330. [PMID: 33775913 PMCID: PMC8328898 DOI: 10.1016/j.jtos.2021.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023]
Abstract
Mucins are a family of high molecular weight, heavily-glycosylated proteins produced by wet epithelial tissues, including the ocular surface epithelia. Densely-packed O-linked glycan chains added post-translationally confer the biophysical properties of hydration, lubrication, anti-adhesion and repulsion. Membrane-associated mucins (MAMs) are the distinguishing components of the mucosal glycocalyx. At the ocular surface, MAMs maintain wetness, lubricate the blink, stabilize the tear film, and create a physical barrier to the outside world. In addition, it is increasingly appreciated that MAMs function as cell surface receptors that transduce information from the outside to the inside of the cell. Recently, our team published a comprehensive review/perspectives article for molecular scientists on ocular surface MAMs, including previously unpublished data and analyses on two new genes MUC21 and MUC22, as well as new MAM functions and biological roles, comparing human and mouse (PMID: 31493487). The current article is a refocus for the audience of The Ocular Surface. First, we update the gene and protein information in a more concise form, and include a new section on glycosylation. Next, we discuss biological roles, with some new sections and further updating from our previous review. Finally, we provide a new chapter on MAM involvement in ocular surface disease. We end this with discussion of an emerging mechanism responsible for damage to the epithelia and their mucosal glycocalyces: the unfolded protein response (UPR). The UPR offers a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Rafael Martinez-Carrasco
- Department of Ophthalmology, Tufts University School of Medicine at New England Eye Center, Tufts Medical Center, Boston, MA, 02111, USA.
| | - Pablo Argüeso
- Department of Ophthalmology, Harvard Medical School at Schepens Eye Research Institute of Mass, Eye and Ear, Boston, MA, 02114, USA.
| | - M Elizabeth Fini
- Department of Ophthalmology, Tufts University School of Medicine at New England Eye Center, Tufts Medical Center: Program in Pharmacology & Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, O2111, USA.
| |
Collapse
|
14
|
Zhu HL, Zhao XW, Han RW, DU QJ, Qi YX, Jiang HN, Huang DW, Yang YX. Changes in bacterial community and expression of genes involved in intestinal innate immunity in the jejunum of newborn lambs during the first 24 hours of life. J Dairy Sci 2021; 104:9263-9275. [PMID: 33985780 DOI: 10.3168/jds.2020-19888] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/29/2021] [Indexed: 12/15/2022]
Abstract
The newborn gut undergoes rapid colonization by commensal microorganisms and possible exposure to pathogens. The contribution of colostrum intake to host protection is well known; however, limited research exists on the intestinal innate immunity corresponding to colostrum intake during the passive immune transfer period in newborn ruminants. The aim of this study was to investigate the changes in bacterial community and expression of genes encoding toll-like receptors (TLR), mucins (MUC), antimicrobial peptides, and tight junctions in the jejunum of lambs that were fed colostrum during the first 24 h of life. Twenty-seven newborn lambs were used in this study, of which 18 lambs were bottle-fed pooled bovine colostrum within the first 2 h after birth to obtain an intake of approximately 8% of body weight. Lambs were slaughtered at 12 (n = 9) and 24 h (n = 9) after birth. The remaining 9 lambs without any feeding were slaughtered at 30 min after birth (0 h). Tissue and ligated segment samples from the jejunum were collected immediately after the lambs were slaughtered. The bacterial profile in the ligated jejunum segment was assessed using amplicon sequencing. The gene expression in the jejunum tissue was determined using quantitative real-time PCR. The relative abundances of Escherichia-Shigella, Lactobacillus, Lactococcus, and Streptococcus increased, whereas those of Sphingomonas, Phyllobacterium, Bradyrhizobium, and Rudaea decreased during the first 24 h of life. Expression of TLR2 and β-defensin 109-like was upregulated at 12 h after birth, but a recovery was detected at 24 h; TLR3, TLR5, LYZ, MUC1, MUC13, MUC20, and CLDN7 showed a higher expression level in samples taken at 24 h than in those taken at 0 h. In addition, expression level of CLDN1, CLDN4, and the junctional adhesion molecule-1 tended to be higher at 24 h than at 0 h after birth. Correlation analysis indicated that TLR2 expression was negatively correlated with the relative abundance of Lactobacillus and Bradyrhizobium, whereas TLR5 expression was positively correlated with the relative abundance of Escherichia-Shigella and Pelagibacterium. These results suggest that TLR, MUC, antimicrobial peptides, and CLDN act together and play an important role in intestinal defense during the passive immune transfer period. They are potentially associated with microbial colonization. The findings from this study provide novel information to elucidate the role of colostrum components in regulating the development of the intestinal mucosal immune barrier in newborn lambs during the passive immune transfer period.
Collapse
Affiliation(s)
- H L Zhu
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China
| | - X W Zhao
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - R W Han
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Q J DU
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - Y X Qi
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - H N Jiang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China
| | - D W Huang
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Y X Yang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China.
| |
Collapse
|
15
|
Yi Q, Liu J, Zhang Y, Qiao H, Chen F, Zhang S, Guan W. Anethole Attenuates Enterotoxigenic Escherichia coli-Induced Intestinal Barrier Disruption and Intestinal Inflammation via Modification of TLR Signaling and Intestinal Microbiota. Front Microbiol 2021; 12:647242. [PMID: 33841372 PMCID: PMC8027122 DOI: 10.3389/fmicb.2021.647242] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the effects of dietary anethole supplementation on the growth performance, intestinal barrier function, inflammatory response, and intestinal microbiota of piglets challenged with enterotoxigenic Escherichia coli K88. Thirty-six weaned piglets (24 ± 1 days old) were randomly allocated into four treatment groups: (1) sham challenge (CON); (2) Escherichia coli K88 challenge (ETEC); (3) Escherichia coli K88 challenge + antibiotics (ATB); and (4) Escherichia coli K88 challenge + anethole (AN). On day 12, the piglets in the ETEC, ATB, and AN group were challenged with 10 mL E. coli K88 (5 × 109 CFU/mL), whereas the piglets in the CON group were orally injected with 10 mL nutrient broth. On day 19, all the piglets were euthanized for sample collection. The results showed that the feed conversion ratio (FCR) was increased in the Escherichia coli K88-challenged piglets, which was reversed by the administration of antibiotics or anethole (P < 0.05). The duodenum and jejunum of the piglets in ETEC group exhibited greater villous atrophy and intestinal morphology disruption than those of the piglets in CON, ATB, and AN groups (P < 0.05). Administration of anethole protected intestinal barrier function and upregulated mucosal layer (mRNA expression of mucin-1 in the jejunum) and tight junction proteins (protein abundance of ZO-1 and Claudin-1 in the ileum) of the piglets challenged with Escherichia coli K88 (P < 0.05). In addition, administration of antibiotics or anethole numerically reduced the plasma concentrations of IL-1β and TNF-α (P < 0.1) and decreased the mRNA expression of TLR5, TLR9, MyD88, IL-1β, TNF-α, IL-6, and IL-10 in the jejunum of the piglets after challenge with Escherichia coli K88 (P < 0.05). Dietary anethole supplementation enriched the abundance of beneficial flora in the intestines of the piglets. In summary, anethole can improve the growth performance of weaned piglets infected by ETEC through attenuating intestinal barrier disruption and intestinal inflammation.
Collapse
Affiliation(s)
- Qingyuan Yi
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiaxin Liu
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yufeng Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hanzhen Qiao
- College of Biological Engineering, Henan University of Technology, Zhengzhou, China
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China.,College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| |
Collapse
|
16
|
Mavi AK, Spalgais S, Singh K, Kumar U, Kumar R. Expression profile of MUC1 protein in Pigeon allergens positive asthmatic. Immunobiology 2021; 226:152086. [PMID: 33784545 DOI: 10.1016/j.imbio.2021.152086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/12/2021] [Accepted: 03/10/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Certain urban areas could contain many pigeon's allergens, which may play an imperative role in the exacerbation of asthma in pigeon allergen sensitive asthma patients. The circulating form of MUC1 in human serum has been considered as a biomarker for some allergic diseases. The study aimed to investigate the role of MUC1 in pigeon allergens positive asthma patients. METHODS We were enrolled 200 asthma patients including 81 males and 119 females. After positive pigeon exposure history, 108 patients underwent SPT testing against pigeon allergens (dropping and feather). A total of 17 patients, who had exposure history with SPT positive were undergone detail clinical examination. Serum MUC1expression analysis was done by western blotting method. RESULTS Out of 200 asthmatic patients, 108 (54%) patients had a history of exposure to pigeons. Skin prick test against pigeon (feather & dropping) allergens was positive in 17 (15.7%) patients among exposure asthmatics. The mean age of the study population was 28.8 ± 10.4 years with 9 males and 8 females. Baseline airway obstruction was seen in 58.8% cases. Out of 17 pigeons expose and sensitive asthmatic the MUC1 expression was up-regulated in 15 (88.2%) and down-regulated in 2 (11.8%). The mean value MUC1 fold change of 15 patients with up-regulation was 4.63 ± 3.00 fold. CONCLUSION MUC1 expression was up-regulated in 88.2% of patients, who were exposed and sensitive to pigeon allergen (dropping and feather). MUC1 may consider as a biomarker in pigeon sensitive asthma patients.
Collapse
Affiliation(s)
- Anil Kumar Mavi
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110007, India
| | - Sonam Spalgais
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110007, India
| | - Kamal Singh
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110007, India
| | - Umesh Kumar
- School of Biosciences, IMS Ghaziabad, NH9 Ghaziabad 201015, Uttar Pradesh, India
| | - Raj Kumar
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110007, India.
| |
Collapse
|
17
|
Lu W, Liu X, Wang T, Liu F, Zhu A, Lin Y, Luo J, Ye F, He J, Zhao J, Li Y, Zhong N. Elevated MUC1 and MUC5AC mucin protein levels in airway mucus of critical ill COVID-19 patients. J Med Virol 2021; 93:582-584. [PMID: 32776556 PMCID: PMC7436726 DOI: 10.1002/jmv.26406] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Wenju Lu
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
| | - Xiaoqing Liu
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
- Department of Critical Care MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Tao Wang
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
| | - Fei Liu
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
| | - Airu Zhu
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
| | - Yongping Lin
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
| | | | - Feng Ye
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
| | - Jianxing He
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
| | - Jincun Zhao
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
| | - Yiming Li
- Division of Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory HealthGuangzhouGuangdongChina
- Department of Critical Care MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Nanshan Zhong
- Department of Critical Care MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
18
|
Hartl K, Sigal M. Microbe-Driven Genotoxicity in Gastrointestinal Carcinogenesis. Int J Mol Sci 2020; 21:E7439. [PMID: 33050171 PMCID: PMC7587957 DOI: 10.3390/ijms21207439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/12/2022] Open
Abstract
The intestinal epithelium serves as a barrier to discriminate the outside from the inside and is in constant exchange with the luminal contents, including nutrients and the microbiota. Pathogens have evolved mechanisms to overcome the multiple ways of defense in the mucosa, while several members of the microbiota can exhibit pathogenic features once the healthy barrier integrity of the epithelium is disrupted. This not only leads to symptoms accompanying the acute infection but may also contribute to long-term injuries such as genomic instability, which is linked to mutations and cancer. While for Helicobacter pylori a link between infection and cancer is well established, many other bacteria and their virulence factors have only recently been linked to gastrointestinal malignancies through epidemiological as well as mechanistic studies. This review will focus on those pathogens and members of the microbiota that have been linked to genotoxicity in the context of gastric or colorectal cancer. We will address the mechanisms by which such bacteria establish contact with the gastrointestinal epithelium-either via an existing breach in the barrier or via their own virulence factors as well as the mechanisms by which they interfere with host genomic integrity.
Collapse
Affiliation(s)
- Kimberly Hartl
- Medical Department, Division of Gastroenterology and Hepatology, Charité-Universtitätsmedizin Berlin, 10117 Berlin, Germany;
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| | - Michael Sigal
- Medical Department, Division of Gastroenterology and Hepatology, Charité-Universtitätsmedizin Berlin, 10117 Berlin, Germany;
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| |
Collapse
|
19
|
Kennel C, Gould EA, Larson ED, Salcedo E, Vickery T, Restrepo D, Ramakrishnan VR. Differential Expression of Mucins in Murine Olfactory Versus Respiratory Epithelium. Chem Senses 2020; 44:511-521. [PMID: 31300812 DOI: 10.1093/chemse/bjz046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mucins are a key component of the surface mucus overlying airway epithelium. Given the different functions of the olfactory and respiratory epithelia, we hypothesized that mucins would be differentially expressed between these 2 areas. Secondarily, we evaluated for potential changes in mucin expression with radiation exposure, given the clinical observations of nasal dryness, altered mucus rheology, and smell loss in radiated patients. Immunofluorescence staining was performed to evaluate expression of mucins 1, 2, 5AC, and 5B in nasal respiratory and olfactory epithelia of control mice and 1 week after exposure to 8 Gy of radiation. Mucins 1, 5AC, and 5B exhibited differential expression patterns between olfactory and respiratory epithelium (RE) while mucin 2 showed no difference. In the olfactory epithelium (OE), mucin 1 was located in a lattice-like pattern around gaps corresponding to dendritic knobs of olfactory sensory neurons, whereas in RE it was intermittently expressed by surface goblet cells. Mucin 5AC was expressed by subepithelial glands in both epithelial types but to a higher degree in the OE. Mucin 5B was expressed by submucosal glands in OE and by surface epithelial cells in RE. At 1-week after exposure to single-dose 8 Gy of radiation, no qualitative effects were seen on mucin expression. Our findings demonstrate that murine OE and RE express mucins differently, and characteristic patterns of mucins 1, 5AC, and 5B can be used to define the underlying epithelium. Radiation (8 Gy) does not appear to affect mucin expression at 1 week. LEVEL OF EVIDENCE N/A (Basic Science Research).IACUC-approved study [Protocol 200065].
Collapse
Affiliation(s)
- Christopher Kennel
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Elizabeth A Gould
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.,Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA.,Neuroscience Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Eric D Larson
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ernesto Salcedo
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Thad Vickery
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.,Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA.,Neuroscience Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Vijay R Ramakrishnan
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA.,Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Neurosurgery, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
20
|
Sheng YH, Ng GZ, Summers KM, Every AL, Price G, Hasnain SZ, Sutton P, McGuckin MA. Influence of the MUC1 Cell Surface Mucin on Gastric Mucosal Gene Expression Profiles in Response to Helicobacter pylori Infection in Mice. Front Cell Infect Microbiol 2020; 10:343. [PMID: 32793510 PMCID: PMC7393270 DOI: 10.3389/fcimb.2020.00343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/04/2020] [Indexed: 11/26/2022] Open
Abstract
The cell surface mucin MUC1 is an important host factor limiting Helicobacter pylori (H. pylori) pathogenesis in both humans and mice by providing a protective barrier and modulating mucosal epithelial and leukocyte responses. The aim of this study was to establish the time-course of molecular events in MUC1-modulated gene expression profiles in response to H. pylori infection in wild type (WT) and MUC1-deficient mice using microarray-determined mRNA expression, gene network analysis and Ingenuity Pathway Analysis (IPA). A time-course over the first 72 h of infection showed significantly higher mucosal loads of bacteria at 8 h of infection in Muc1−/− mice compared with WT, confirming its importance in the early stages of infection (P = 0.0003). Microarray analysis revealed 266 differentially expressed genes at one or more time-points over 72 h in the gastric mucosa of Muc1−/− mice compared with WT control using a threshold of 2-fold change. The SPINK1 pancreatic cancer canonical pathway was strongly inhibited in Muc1−/− mice compared with WT at sham and 8 h infection (P = 6.08E-14 and P = 2.25 E-19, respectively) but potently activated at 24 and 72 h post-infection (P = 1.38E-22 and P = 5.87E-13, respectively). The changes in this pathway are reflective of higher expression of genes mediating digestion and absorption of lipids, carbohydrates, and proteins at sham and 8 h infection in the absence of MUC1, but that this transcriptional signature is highly down regulated as infection progresses in the absence of MUC1. Uninfected Muc1−/− gastric tissue was highly enriched for expression of factors involved in lipid metabolism and 8 h infection further activated this network compared with WT. As infection progressed, a network of antimicrobial and anti-inflammatory response genes was more highly activated in Muc1−/− than WT mice. Key target genes identified by time-course microarrays were independently validated using RT-qPCR. These results highlight the dynamic interplay between the host and H. pylori, and the role of MUC1 in host defense, and provide a general picture of changes in cellular gene expression modulated by MUC1 in a time-dependent manner in response to H. pylori infection.
Collapse
Affiliation(s)
- Yong H Sheng
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Garrett Z Ng
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Kim M Summers
- Genetics, Genomics & Transcriptomics of Disease Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Alison L Every
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Gareth Price
- QCIF Facility for Advanced Bioinformatics, Institute of Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Sumaira Z Hasnain
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Philip Sutton
- Mucosal Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, Faculty of Medicine Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Michael A McGuckin
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia.,Faculty of Medicine Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
21
|
Wang YM, Qi X, Gong FC, Chen Y, Yang ZT, Mao EQ, Chen EZ. Protective and predictive role of Mucin1 in sepsis-induced ALI/ARDS. Int Immunopharmacol 2020; 83:106438. [PMID: 32247267 DOI: 10.1016/j.intimp.2020.106438] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVE We aimed to investigate whether inhibition of MUC1 would aggravate sepsis-induced ALI, and explore the predictive value of plasma MUC1 for sepsis patients with or without ARDS. MATERIALS AND METHODS MUC1 siRNA pre-treatment was used to knockdown MUC1 expression in vitro. GO203 was used to inhibit the homodimerization of MUC1-C in vivo. Expression levels of MUC1, TLR 4 and HIF-1α were detected by Western blot. In addition, plasma MUC1 levels of enrolled patients were detected by ELISA on the day of admission and on the 3rd day. ROC curve was used to determine the predictive value of MUC1 in sepsis patients with ARDS. RESULTS Our results showed that inhibition of MUC1 could aggravate sepsis-induced acute lung injury and increase the expression of inflammatory cytokines in sera and BALF of sepsis mice. At the same time, we confirmed that inhibition of MUC1 could significantly decrease HIF-1α expression and thereby activate the expression level of TLR4. HIF-1α was a negative regulator of TLR-4. In addition, plasma MUC1 levels of sepsis patients with ARDS were significantly higher than those without ARDS and healthy adults. ROC curve showed that predictive value of plasma MUC1 on sepsis with ARDS on the 3rd day of enrollment was higher than the day of enrollment. CONCLUSION MUC1 could inhibit the expression of TLR-4 by stabilizing HIF-1α, thereby alleviate sepsis-induced lung injury and protect organ function. At the same time, elevated MUC1 levels in plasma had a good predictive valud on whether patients with sepsis would develop ARDS.
Collapse
Affiliation(s)
- Yu-Ming Wang
- Department of Emergency in Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xing Qi
- Department of Emergency in Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Fang-Chen Gong
- Department of Emergency in Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ying Chen
- Department of Emergency in Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Zhi-Tao Yang
- Department of Emergency in Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - En-Qiang Mao
- Department of Emergency in Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Er-Zhen Chen
- Department of Emergency in Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
22
|
Bose M, Mukherjee P. Microbe-MUC1 Crosstalk in Cancer-Associated Infections. Trends Mol Med 2020; 26:324-336. [PMID: 31753595 DOI: 10.1016/j.molmed.2019.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 02/07/2023]
Abstract
Infection-associated cancers account for ∼20% of all malignancies. Understanding the molecular mechanisms underlying infection-associated malignancies may help in developing diagnostic biomarkers and preventative vaccines against malignancy. During infection, invading microbes interact with host mucins lining the glandular epithelial cells and trigger inflammation. MUC1 is a transmembrane mucin glycoprotein that is present on the surface of almost all epithelial cells, and is known to interact with invading microbes. This interaction can trigger pro- or anti-inflammatory responses depending on the microbe and the cell type. In this review we summarize the mechanisms of microbe and MUC1 interactions, and highlight how MUC1 plays contrasting roles in different cells. We also share perspectives on future research that may support clinical advances in infection-associated cancers.
Collapse
Affiliation(s)
- Mukulika Bose
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA.
| | - Pinku Mukherjee
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
23
|
Muc5ac null mice are predisposed to spontaneous gastric antro-pyloric hyperplasia and adenomas coupled with attenuated H. pylori-induced corpus mucous metaplasia. J Transl Med 2019; 99:1887-1905. [PMID: 31399638 PMCID: PMC6927550 DOI: 10.1038/s41374-019-0293-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide and is strongly associated with chronic Helicobacter pylori (Hp) infection. The ability of Hp to closely adhere to the gastric surface protective mucous layer containing mucins (MUC in humans and Muc in animals), primarily Muc5ac, is integral in the stepwise pathogenesis from gastritis to cancer. To probe the role of Muc5ac in Hp-induced gastric pathology, Muc5ac-/- and Muc5ac+/+ (WT) mice were experimentally infected with Hp Sydney strain (SS1). At 16 weeks and 32 weeks post infection (wpi), groups of mice were euthanized and evaluated for the following: gastric histopathological parameters, immunohistochemical expression of mucins (Muc5ac, Muc1, Muc2), Trefoil factor family proteins (Tff1 and Tff2), Griffonia (Bandeiraea) simplicifolia lectin II (GSL II) (mucous metaplasia marker) and Clusterin (Spasmolytic Polypeptide Expressing Metaplasia (SPEM) marker), Hp colonization density by qPCR and gastric cytokine mRNA levels. Our results demonstrate that Muc5ac-/- mice developed spontaneous antro-pyloric proliferation, adenomas and in one case with neuroendocrine differentiation; these findings were independent of Hp infection along with strong expression levels of Tff1, Tff2 and Muc1. Hp-infected Muc5ac-/- mice had significantly lowered gastric corpus mucous metaplasia at 16 wpi and 32 wpi (P = 0.0057 and P = 0.0016, respectively), with a slight reduction in overall gastric corpus pathology. GSII-positive mucous neck cells were decreased in Hp-infected Muc5ac-/- mice compared to WT mice and clusterin positivity was noted within metaplastic glands in both genotypes following Hp infection. Additionally, Hp colonization densities were significantly higher in Muc5ac-/- mice compared to WT at 16 wpi in both sexes (P = 0.05) along with a significant reduction in gastric Tnfα (16 wpi-males and females, P = 0.017 and P = 0.036, respectively and 32 wpi-males only, P = 0.025) and Il-17a (16 wpi-males) (P = 0.025). Taken together, our findings suggest a protective role for MUC5AC/Muc5ac in maintaining gastric antral equilibrium and inhibiting Hp colonization and associated inflammatory pathology.
Collapse
|
24
|
Walduck AK, Raghavan S. Immunity and Vaccine Development Against Helicobacter pylori. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1149:257-275. [PMID: 31016627 DOI: 10.1007/5584_2019_370] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Helicobacter pylori is a highly-adapted gastrointestinal pathogen of humans and the immunology of this chronic infection is extremely complex. Despite the availability of antibiotic therapy, the global incidence of H. pylori infection remains high, particularly in low to middle-income nations. Failure of therapy and the spread of antibiotic resistance among the bacteria are significant problems and provide impetus for the development of new therapies and vaccines to treat or prevent gastric ulcer, and gastric carcinoma. The expansion of knowledge on gastric conventional and regulatory T cell responses, and the role of TH17 in chronic gastritis from studies in mouse models and patients have provided valuable insights into how gastritis is initiated and maintained. The development of human challenge models for testing candidate vaccines has meant a unique opportunity to study acute infection, but the field of vaccine development has not progressed as rapidly as anticipated. One clear lesson learned from previous studies is that we need a better understanding of the immune suppressive mechanisms in vivo to be able to design vaccine strategies. There is still an urgent need to identify practical surrogate markers of protection that could be deployed in future field vaccine trials. Important developments in our understanding of the chronic inflammatory response, progress and problems arising from human studies, and an outlook for the future of clinical vaccine trials will be discussed.
Collapse
Affiliation(s)
- Anna K Walduck
- School of Science, RMIT University, Melbourne, VIC, Australia.
| | - Sukanya Raghavan
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
25
|
Sharma L, Rebaza A, Dela Cruz CS. When "B" becomes "A": the emerging threat of influenza B virus. Eur Respir J 2019; 54:54/2/1901325. [PMID: 31416813 DOI: 10.1183/13993003.01325-2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 11/05/2022]
Affiliation(s)
- Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Andre Rebaza
- Section of Pediatric Pulmonary, Allergy, Immunology and Sleep Medicine, Dept of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Dept of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
26
|
Dhar P, McAuley J. The Role of the Cell Surface Mucin MUC1 as a Barrier to Infection and Regulator of Inflammation. Front Cell Infect Microbiol 2019; 9:117. [PMID: 31069176 PMCID: PMC6491460 DOI: 10.3389/fcimb.2019.00117] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/02/2019] [Indexed: 12/27/2022] Open
Abstract
The family of cell surface (cs-) mucins are constitutively expressed at the cell surface by nearly all epithelial cells, beneath the gel-mucin layer. All cs-mucin family members have structural features that enable them to act as a releasable decoy barrier to mucosal pathogens, by providing ligands for pathogen binding and the ability to shed the bound extracellular domain. Due to the towering structure of cs-mucins at the surface, binding of mucosal pathogens can also sterically block binding to underlying cellular receptors. The cytoplasmic tail domain of cs-mucins are capable of initiating signal transduction cascades and due to their conservation across species, may play an important biological role in cellular signaling. MUC1 is one of the most extensively studied of the cs-mucin family. With respect to its physiological function in the mucosal environment, MUC1 has been demonstrated to play a dynamic role in protection of the host from infection by a wide variety of pathogens and to regulate inflammatory responses to infection. This review briefly summarizes the current knowledge and new findings regarding the structural features relating to the function of MUC1, its role as a protective barrier against pathogen invasion and mechanisms by which this cs-mucin regulates inflammation.
Collapse
Affiliation(s)
- Poshmaal Dhar
- Faculty of Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Julie McAuley
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Yoon JH, Ham IH, Kim O, Ashktorab H, Smoot DT, Nam SW, Lee JY, Hur H, Park WS. Gastrokine 1 protein is a potential theragnostic target for gastric cancer. Gastric Cancer 2018; 21:956-967. [PMID: 29704153 DOI: 10.1007/s10120-018-0828-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/19/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gastrokine 1 (GKN1) plays important roles in maintaining mucosal homeostasis, and in regulating cell proliferation and differentiation. Here, we determined whether GKN1 is a potential theragnostic marker for gastric cancer. METHODS We identified GKN1 binding proteins using the protein microarray assay and investigated whether GKN1 is one of the exosomal cargo proteins by western blot, immunoprecipitation, and immunofluorescent assays. Cell proliferation and apoptosis were analyzed by MTT, BrdU incorporation, flow cytometry, and western blot assays. We further validated the functional relevance of exosomal GKN1 in MKN1-injected xenograft mice. The possibility of serum GKN1 as a diagnostic marker for gastric cancer was determined by ELISA assay. RESULTS In protein microarray assay, GKN1 binding to 27 exosomal proteins was clearly observed. GKN1 was expressed in exosomes derived from HFE-145 gastric epithelial cells by western blot and immunofluorescent assays, but not in exosomes from AGS and MKN1 gastric cancer cells. Exosomes carrying GKN1 inhibited cell proliferation and induced apoptosis in both AGS and MKN1 cells, and exosomes carrying GKN1-treated nude mice-bearing MKN1 xenograft tumors exhibited significantly reduced tumor volume and tumor weight. Silencing of clathrin markedly down-regulated the internalization of exosomal GKN1. Interestingly, serum GKN1 concentrations in patients with gastric cancer were significantly lower than those in healthy individuals and patients with colorectal and hepatocellular carcinomas. CONCLUSIONS The GKN1 is secreted and internalized in the gastric epithelium by exosome-driven transfer, which inhibits gastric tumorigenesis and supports the clinical application of GKN1 protein in gastric cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
- Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - In-Hye Ham
- Department of Surgery, Ajou University School of Medicine, Suwon, 16499, South Korea
- Brain Korea 21 Plus Research Center for Biomedical Science, Ajou University, Suwon, 16499, South Korea
| | - Olga Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - Hassan Ashktorab
- Department of Medicine, Howard University, Washington, DC, 20060, USA
| | - Duane T Smoot
- Department of Medicine, Meharry Medical Center, Nashville, TN, 37208, USA
| | - Suk Woo Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
- Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - Jung Young Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
- Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, 16499, South Korea.
- Brain Korea 21 Plus Research Center for Biomedical Science, Ajou University, Suwon, 16499, South Korea.
| | - Won Sang Park
- Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.
- Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.
| |
Collapse
|
28
|
Radziejewska I, Borzym-Kluczyk M, Leszczyńska K, Wosek J, Bielawska A. Lotus tetragonolobus and Maackia amurensis lectins influence phospho-IκBα, IL-8, Lewis b and H type 1 glycoforms levels in H. pylori infected CRL-1739 gastric cancer cells. Adv Med Sci 2018; 63:205-211. [PMID: 29197783 DOI: 10.1016/j.advms.2017.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/01/2017] [Accepted: 06/13/2017] [Indexed: 12/20/2022]
Abstract
PURPOSE Attachment of Helicobacter pylori to the mucous epithelial cells and the mucous layer is said to be a crucial step for infection development. Sugar antigens of gastric mucins (MUC5AC, MUC1) can act as receptors for bacterial adhesins. The aim of the study was to investigate if Lotus tetragonolobus and Maackia amurensis lectins influence the level of MUC1, MUC5AC, Lewis b, H type 1, sialyl Lewis x, phospho-IκBα and interleukin 8 in Helicobacter pylori infected gastric cancer cells. MATERIALS AND METHODS The study was performed with one clinical H. pylori strain and CRL-1739 gastric cancer cells. To assess the levels of mentioned factors immunosorbent ELISA assays were used. RESULTS Coculture of cells with bacteria had no clear effect on almost all examined structures. After coculture with H. pylori and lectins, a decrease of the level of both mucins, Lewis b and H type 1 antigens was observed. Lectins addition had no effect on sialyl Lewis x. Maackia amurensis caused slight increase of phospho-IκBα while interleukin 8 level was decreased. CONCLUSIONS Lotus tetragonolobus and Maackia amurensis lectins can mediate in binding of Helicobacter pylori to gastric epithelium.
Collapse
|
29
|
Lock JY, Carlson TL, Carrier RL. Mucus models to evaluate the diffusion of drugs and particles. Adv Drug Deliv Rev 2018; 124:34-49. [PMID: 29117512 DOI: 10.1016/j.addr.2017.11.001] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/12/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022]
Abstract
Mucus is a complex hydrogel that acts as a natural barrier to drug delivery at different mucosal surfaces including the respiratory, gastrointestinal, and vaginal tracts. To elucidate the role mucus plays in drug delivery, different in vitro, in vivo, and ex vivo mucus models and techniques have been utilized. Drug and drug carrier diffusion can be studied using various techniques in either isolated mucus gels or mucus present on cell cultures and tissues. The species, age, and potential disease state of the animal from which mucus is derived can all impact mucus composition and structure, and therefore impact drug and drug carrier diffusion. This review provides an overview of the techniques used to characterize drug and drug carrier diffusion, and discusses the advantages and disadvantages of the different models available to highlight the information they can afford.
Collapse
|
30
|
MUC1: The First Respiratory Mucin with an Anti-Inflammatory Function. J Clin Med 2017; 6:jcm6120110. [PMID: 29186029 PMCID: PMC5742799 DOI: 10.3390/jcm6120110] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023] Open
Abstract
MUC1 is a membrane-bound mucin expressed on the apical surfaces of most mucosal epithelial cells. In normal lung epithelia, MUC1 is a binding site for Pseudomonas aeruginosa, an opportunistic human pathogen of great clinical importance. It has now been established that MUC1 also serves an anti-inflammatory role in the airways that is initiated late in the course of a bacterial infection and is mediated through inhibition of Toll-like receptor (TLR) signaling. MUC1 expression was initially shown to interfere with TLR5 signaling in response to P. aeruginosa flagellin, but has since been extended to other TLRs. These new findings point to an immunomodulatory role for MUC1 during P. aeruginosa lung infection, particularly during the resolution phase of inflammation. This review briefly summarizes the recent characterization of MUC1’s anti-inflammatory properties in both the respiratory tract and extrapulmonary tissues.
Collapse
|
31
|
Kim O, Yoon JH, Choi WS, Ashktorab H, Smoot DT, Nam SW, Lee JY, Park WS. Heterodimeric interaction between GKN2 and TFF1 entails synergistic antiproliferative and pro-apoptotic effects on gastric cancer cells. Gastric Cancer 2017; 20:772-783. [PMID: 28150071 PMCID: PMC5718056 DOI: 10.1007/s10120-017-0692-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 01/14/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND GKN2 and TFF1 form a heterodimer that is only generated in the mucus-secreting cells of the normal stomach. The formation of this heterodimer is frequently disrupted in gastric cancer. However, the precise roles of GKN2 alone and in the heterodimer with TFF1 as well as the contributions of GKN2 and the heterodimer to gastric carcinogenesis are poorly understood. METHODS Cell viability, proliferation, and apoptosis were analyzed in AGS, MKN1, MKN28, and MKN45 gastric cancer cells transfected with GKN2 and/or TFF1 using MTT, BrdU incorporation, and apoptosis assays, respectively. In addition, cell viability was examined in HFE-145 non-neoplastic gastric epithelial cells after GKN2 and/or TFF1 silencing. Furthermore, the cell cycle and the expression of cell cycle and apoptosis related proteins were assessed. The interaction between GKN2 and TFF1 was confirmed by co-immunoprecipitation. Immunohistochemistry was employed to explore TFF1 expression in 169 gastric cancer tissues. RESULTS Co-transfection with GKN2 and TFF1 significantly inhibited cell viability and proliferation by inducing G1/S cell cycle arrest and suppressing positive cell cycle regulators. Simultaneous knockdown of GKN2 and TFF1 in HFE-145 cells resulted in markedly increased cell viability. Moreover, the interaction of GKN2 and TFF1 promoted cell death by enhancing caspase-3/7 activity and upregulating pro-apoptotic proteins. At the mRNA level, GKN2 and TFF1 were found to be positively correlated in non-tumor and tumor samples. Immunohistochemistry revealed loss of TFF1 expression in 128 (75.73%) of 169 gastric cancers. There was a borderline-significant association between GKN2 and TFF1 protein expression in gastric cancers (P = 0.0598). CONCLUSION Collectively, our data demonstrated that the interaction between GKN2 and TFF1 can have synergistic antiproliferative and pro-apoptotic effects on gastric cancer.
Collapse
Affiliation(s)
- Olga Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Korea
| | - Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Korea
| | - Won Suk Choi
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Korea
| | - Hassan Ashktorab
- Department of Medicine, Howard University, Washington, DC, 20060, USA
| | - Duane T Smoot
- Department of Medicine, Howard University, Washington, DC, 20060, USA
| | - Suk Woo Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Korea
| | - Jung Young Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Korea
| | - Won Sang Park
- Department of Pathology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Korea.
| |
Collapse
|
32
|
van Putten JPM, Strijbis K. Transmembrane Mucins: Signaling Receptors at the Intersection of Inflammation and Cancer. J Innate Immun 2017; 9:281-299. [PMID: 28052300 DOI: 10.1159/000453594] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 11/19/2016] [Indexed: 12/18/2022] Open
Abstract
Mucosal surfaces line our body cavities and provide the interaction surface between commensal and pathogenic microbiota and the host. The barrier function of the mucosal layer is largely maintained by gel-forming mucin proteins that are secreted by goblet cells. In addition, mucosal epithelial cells express cell-bound mucins that have both barrier and signaling functions. The family of transmembrane mucins consists of diverse members that share a few characteristics. The highly glycosylated extracellular mucin domains inhibit invasion by pathogenic bacteria and can form a tight mesh structure that protects cells in harmful conditions. The intracellular tails of transmembrane mucins can be phosphorylated and connect to signaling pathways that regulate inflammation, cell-cell interactions, differentiation, and apoptosis. Transmembrane mucins play important roles in preventing infection at mucosal surfaces, but are also renowned for their contributions to the development, progression, and metastasis of adenocarcinomas. In general, transmembrane mucins seem to have evolved to monitor and repair damaged epithelia, but these functions can be highjacked by cancer cells to yield a survival advantage. This review presents an overview of the current knowledge of the functions of transmembrane mucins in inflammatory processes and carcinogenesis in order to better understand the diverse functions of these multifunctional proteins.
Collapse
Affiliation(s)
- Jos P M van Putten
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
33
|
Kato K, Uchino R, Lillehoj EP, Knox K, Lin Y, Kim KC. Membrane-Tethered MUC1 Mucin Counter-Regulates the Phagocytic Activity of Macrophages. Am J Respir Cell Mol Biol 2016; 54:515-23. [PMID: 26393683 DOI: 10.1165/rcmb.2015-0177oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
MUC1 (MUC in human; Muc in animals) is a transmembrane mucin glycoprotein expressed in mucosal epithelial cells and hematopoietic cells. MUC1 is involved in the resolution of inflammation during airway Pseudomonas aeruginosa (Pa) infection by suppressing Toll-like receptor signaling in airway epithelial cells. Although alveolar macrophages are recognized as critical mediators of cell-mediated immunity against microorganisms inhaled into the airways, the role of MUC1 in regulating their response is unknown. The aims of this study were to determine whether macrophages express MUC1, and, if so, whether MUC1 expression might be associated with macrophage M0/M1/M2 differentiation or phagocytic activity. Human and mouse MUC1/Muc1 expression was drastically up-regulated in classically activated (M1) macrophages compared with nonactivated (M0) and alternatively activated (M2) macrophages. M1 polarization and Pa stimulation each increased MUC1 ectodomain shedding from the macrophage surface in a TNF-α-converting enzyme-dependent manner. MUC1/Muc1 deficiency in M0 macrophages increased adhesion and phagocytosis of Pa and Escherichia coli compared with MUC1/Muc1-expressing cells, and attenuation of phagocytosis by MUC1 was augmented after polarization into M1 macrophages compared with M0 macrophages. Finally, MUC1/Muc1 deficiency in macrophages increased reactive oxygen species production and TNF-α release in response to Pa compared with MUC1/Muc1-sufficient cells. These results indicate that MUC1/Muc1 expression by macrophages is predominantly in the M1 subtype, and that MUC1/Muc1 expression in these cells decreases their phagocytic activity in an antiinflammatory manner.
Collapse
Affiliation(s)
- Kosuke Kato
- 1 Department of Otolaryngology, University of Arizona College of Medicine, Tucson, Arizona.,2 Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Reina Uchino
- 2 Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Erik P Lillehoj
- 3 Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kenneth Knox
- 4 Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona; and
| | - Yong Lin
- 5 Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico(Received in original form May 28, 2015 and in final form September 15, 2015)
| | - K Chul Kim
- 1 Department of Otolaryngology, University of Arizona College of Medicine, Tucson, Arizona.,2 Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Regulation of Airway Inflammation by G-protein Regulatory Motif Peptides of AGS3 protein. Sci Rep 2016; 6:27054. [PMID: 27270970 PMCID: PMC4895231 DOI: 10.1038/srep27054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 05/12/2016] [Indexed: 12/21/2022] Open
Abstract
Respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), and lung infections have critical consequences on mortality and morbidity in humans. The aims of the present study were to examine the mechanisms by which CXCL12 affects MUC1 transcription and airway inflammation, which depend on activator of G-protein signaling (AGS) 3 and to identify specific molecules that suppress CXCL12-induced airway inflammation by acting on G-protein-coupled receptors. Herein, AGS3 suppresses CXCL12-mediated upregulation of MUC1 and TNFα by regulating Gαi. We found that the G-protein regulatory (GPR) motif peptide in AGS3 binds to Gαi and downregulates MUC1 expression; in contrast, this motif upregulates TNFα expression. Mutated GPR Q34A peptide increased the expression of MUC1 and TGFβ but decreased the expression of TNFα and IL-6. Moreover, CXCR4-induced dendritic extensions in 2D and 3D matrix cultures were inhibited by the GPR Q34A peptide compared with a wild-type GPR peptide. The GPR Q34A peptide also inhibited CXCL12-induced morphological changes and inflammatory cell infiltration in the mouse lung, and production of inflammatory cytokines in bronchoalveolar lavage (BAL) fluid and the lungs. Our data indicate that the GPR motif of AGS3 is critical for regulating MUC1/Muc1 expression and cytokine production in the inflammatory microenvironment.
Collapse
|
35
|
Zhang L, Wu WKK, Gallo RL, Fang EF, Hu W, Ling TKW, Shen J, Chan RLY, Lu L, Luo XM, Li MX, Chan KM, Yu J, Wong VWS, Ng SC, Wong SH, Chan FKL, Sung JJY, Chan MTV, Cho CH. Critical Role of Antimicrobial Peptide Cathelicidin for Controlling Helicobacter pylori Survival and Infection. THE JOURNAL OF IMMUNOLOGY 2016; 196:1799-1809. [DOI: 10.4049/jimmunol.1500021] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
The antimicrobial peptide cathelicidin is critical for protection against different kinds of microbial infection. This study sought to elucidate the protective action of cathelicidin against Helicobacter pylori infection and its associated gastritis. Exogenous cathelicidin was found to inhibit H. pylori growth, destroy the bacteria biofilm, and induce morphological alterations in H. pylori membrane. Additionally, knockdown of endogenous cathelicidin in human gastric epithelial HFE-145 cells markedly increased the intracellular survival of H. pylori. Consistently, cathelicidin knockout mice exhibited stronger H. pylori colonization, higher expression of proinflammatory cytokines IL-6, IL-1β, and ICAM1, and lower expression of the anti-inflammatory cytokine IL-10 in the gastric mucosa upon H. pylori infection. In wild-type mice, H. pylori infection also stimulated gastric epithelium-derived cathelicidin production. Importantly, pretreatment with bioengineered Lactococcus lactis that actively secretes cathelicidin significantly increased mucosal cathelicidin levels and reduced H. pylori infection and the associated inflammation. Moreover, cathelicidin strengthened the barrier function of gastric mucosa by stimulating mucus synthesis. Collectively, these findings indicate that cathelicidin plays a significant role as a potential natural antibiotic for H. pylori clearance and a therapeutic agent for chronic gastritis.
Collapse
Affiliation(s)
- Lin Zhang
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - William K. K. Wu
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
- §Department of Anesthesia and Intensive Care, Chinese University of Hong Kong, Hong Kong, China
| | - Richard L. Gallo
- ¶Division of Dermatology, University of California, San Diego, La Jolla, CA 92093
| | - Evandro F. Fang
- ‖Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224
| | - Wei Hu
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Thomas K. W. Ling
- **Department of Microbiology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong, China
| | - Jing Shen
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Ruby L. Y. Chan
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Lan Lu
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Xiao M. Luo
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Ming X. Li
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Kam M. Chan
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| | - Jun Yu
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - Vincent W. S. Wong
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Sunny H. Wong
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - Francis K. L. Chan
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - Joseph J. Y. Sung
- *Institute of Digestive Diseases and State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- †Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- ‡CUHK Shenzhen Research Institute, Shenzhen 518057, China
| | - Matthew T. V. Chan
- §Department of Anesthesia and Intensive Care, Chinese University of Hong Kong, Hong Kong, China
| | - Chi H. Cho
- #School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; and
| |
Collapse
|
36
|
Shirai K, Saika S. Ocular surface mucins and local inflammation--studies in genetically modified mouse lines. BMC Ophthalmol 2015; 15 Suppl 1:154. [PMID: 26818460 PMCID: PMC4895702 DOI: 10.1186/s12886-015-0137-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Mucins locate to the apical surfaces of all wet-surfaced epithelia including ocular surface. The functions of the mucins include anti-adhesive, lubrication, water retention, allergens and pathogen barrier function. Ocular surface pathologies, i.e. dry eye syndrome or allergic conjunctivitis, are reportedly associated with alteration of expression pattern of mucin components. Recent investigations indicated anti-bacterial adhesion or anti-inflammatory effects of members of mucins in non-ocular tissues, i.e., gastrointestinal tracts or airway tissues, by using genetically modified mouse lines that lacks an expression of a mucin member. However, examination of ocular phenotypes of each of mucin gene-ablated mouse lines has not yet fully performed. Muc16-dficient mouse is associated with spontaneous subclinical inflammation in conjunctiva. The article reviews the roles of mucin members in modulation of local inflammation in mucous membrane tissues and phenotype of mouse lines with the loss of a mucin gene. Analysis of ocular surface of mucin-gene related mutant mouse lines are to be further performed.
Collapse
Affiliation(s)
- Kumi Shirai
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan.
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, Wakayama, 641-0012, Japan.
| |
Collapse
|
37
|
Apostolopoulos V, Stojanovska L, Gargosky SE. MUC1 (CD227): a multi-tasked molecule. Cell Mol Life Sci 2015; 72:4475-500. [PMID: 26294353 PMCID: PMC11113675 DOI: 10.1007/s00018-015-2014-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/23/2015] [Accepted: 08/06/2015] [Indexed: 12/16/2022]
Abstract
Mucin 1 (MUC1 [CD227]) is a high-molecular weight (>400 kDa), type I membrane-tethered glycoprotein that is expressed on epithelial cells and extends far above the glycocalyx. MUC1 is overexpressed and aberrantly glycosylated in adenocarcinomas and in hematological malignancies. As a result, MUC1 has been a target for tumor immunotherapeutic studies in mice and in humans. MUC1 has been shown to have anti-adhesive and immunosuppressive properties, protects against infections, and is involved in the oncogenic process as well as in cell signaling. In addition, MUC1 plays a key role in the reproductive tract, in the immune system (affecting dendritic cells, monocytes, T cells, and B cells), and in chronic inflammatory diseases. Evidence for all of these roles for MUC1 is discussed herein and demonstrates that MUC1 is truly a multitasked molecule.
Collapse
Affiliation(s)
- Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.
| | - Lily Stojanovska
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | | |
Collapse
|
38
|
Menon BB, Kaiser-Marko C, Spurr-Michaud S, Tisdale AS, Gipson IK. Suppression of Toll-like receptor-mediated innate immune responses at the ocular surface by the membrane-associated mucins MUC1 and MUC16. Mucosal Immunol 2015; 8:1000-8. [PMID: 25563498 PMCID: PMC4495011 DOI: 10.1038/mi.2014.127] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 12/01/2014] [Indexed: 02/04/2023]
Abstract
Membrane-associated mucins (MAMs) expressed on the ocular surface epithelium form a dense glycocalyx that is hypothesized to protect the cornea and conjunctiva from external insult. In this study, the hypothesis that the MAMs MUC1 and MUC16, expressed on the apical surface of the corneal epithelium, suppress Toll-like receptor (TLR)-mediated innate immune responses was tested. Using an in vitro model of corneal epithelial cells that are cultured to express MAMs, we show that reduced expression of either MUC1 or MUC16 correlates with increased message and secreted protein levels of the proinflammatory cytokines interleukin (IL)-6, IL-8, and tumor necrosis factor-α (TNF-α) following exposure of cells to the TLR2 and TLR5 agonists, heat-killed Listeria monocytogenes and flagellin, respectively. As mice express Muc1 (but not Muc16) in the corneal epithelium, a Muc1(-/-) mouse model was used to extend in vitro findings. Indeed, IL-6 and TNF-α message levels were increased in the corneal epithelium of Muc1(-/-) mice, in comparison with wild-type mice, following exposure of enucleated eyes to the TLR2 and TLR5 agonists. Our results suggest that the MAMs MUC1 and MUC16 contribute to the maintenance of immune homeostasis at the ocular surface by limiting TLR-mediated innate immune responses.
Collapse
Affiliation(s)
- Balaraj B. Menon
- Address correspondence to: Balaraj B. Menon, Ph.D., Schepens Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford St., Boston, MA 02114, Tel: 617-912-0217,
| | | | | | | | | |
Collapse
|
39
|
Parigi SM, Eldh M, Larssen P, Gabrielsson S, Villablanca EJ. Breast Milk and Solid Food Shaping Intestinal Immunity. Front Immunol 2015; 6:415. [PMID: 26347740 PMCID: PMC4541369 DOI: 10.3389/fimmu.2015.00415] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/28/2015] [Indexed: 12/22/2022] Open
Abstract
After birth, the intestinal immune system enters a critical developmental stage, in which tolerogenic and pro-inflammatory cells emerge to contribute to the overall health of the host. The neonatal health is continuously challenged by microbial colonization and food intake, first in the form of breast milk or formula and later in the form of solid food. The microbiota and dietary compounds shape the newborn immune system, which acquires the ability to induce tolerance against innocuous antigens or induce pro-inflammatory immune responses against pathogens. Disruption of these homeostatic mechanisms might lead to undesired immune reactions, such as food allergies and inflammatory bowel disease. Hence, a proper education and maturation of the intestinal immune system is likely important to maintain life-long intestinal homeostasis. In this review, the most recent literature regarding the effects of dietary compounds in the development of the intestinal immune system are discussed.
Collapse
Affiliation(s)
- Sara M Parigi
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| | - Maria Eldh
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| | - Pia Larssen
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| | - Susanne Gabrielsson
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| | - Eduardo J Villablanca
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| |
Collapse
|
40
|
Cha HJ, Jung MS, Ahn DW, Choi JK, Ock MS, Kim KS, Yoon JH, Song EJ, Song KS. Silencing of MUC8 by siRNA increases P2Y₂-induced airway inflammation. Am J Physiol Lung Cell Mol Physiol 2015; 308:L495-502. [PMID: 25575516 DOI: 10.1152/ajplung.00332.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mucin hypersecretion and overproduction are frequent manifestations of respiratory disease. Determining the physiological function of airway mucin is presently considered more important than identifying the relevant signaling pathways. The lack of a full-length human mucin 8 (MUC8) cDNA sequence has hindered the generation of a Muc8 knockout mouse line. Thus, the precise physiological functions of MUC8 are unclear. Herein, we investigated the function of MUC8 using a small-interfering RNA (siRNA)-mediated genetic silencing approach in human airway epithelial cells. Herein, intracellular IL-1α production was stimulated by an ATP/P2Y2 complex. While ATP/P2Y₂ increased IL-1α secretion in a time-dependent manner, treatment with P2Y₂-specific siRNA significantly decreased IL-1α secretion. Moreover, ATP increased P2Y₂-mediated upregulation of MUC8 expression; however, IL-1α significantly decreased the extent to which ATP/P2Y₂ upregulated MUC8 expression. Interestingly, treatment with MUC8-specific siRNA decreased the production of anti-inflammatory cytokines (TGF-β and IL-1 receptor antagonist) and increased the production of inflammatory cytokines (IL-1α and IL-6) in our system. In addition, siRNA-mediated knockdown of MUC8 expression dramatically increased the secretion of inflammatory chemokines and resulted in an approximately threefold decrease in cell chemotaxis. We propose that MUC8 may function as an anti-inflammatory mucin that participates in inflammatory response by attracting immune cells/cytokines to the site of inflammation. Our results provide new insight into the physiological function of MUC8 and enhance our understanding of mucin overproduction during airway inflammation.
Collapse
Affiliation(s)
- Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan, Korea; Institute of Medicine, Kosin University College of Medicine, Busan, Korea
| | - Min-Su Jung
- Department of Physiology, Kosin University College of Medicine, Busan, Korea
| | - Do Whan Ahn
- Department of Physiology, Kosin University College of Medicine, Busan, Korea
| | - Jang-Kyu Choi
- Department of Physiology, Kosin University College of Medicine, Busan, Korea
| | - Mee Sun Ock
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan, Korea
| | - Kyung Soo Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chung-Ang University School of Medicine, Seoul, Korea
| | - Joo-Heon Yoon
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea; and
| | - Eun Ju Song
- Department of Veterinary Physiology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Kyoung Seob Song
- Institute of Medicine, Kosin University College of Medicine, Busan, Korea; Department of Physiology, Kosin University College of Medicine, Busan, Korea
| |
Collapse
|
41
|
|
42
|
Yao F, Liu H, Li Z, Zhong C, Fang W. Down-regulation of LATS2 in non-small cell lung cancer promoted the growth and motility of cancer cells. Tumour Biol 2014; 36:2049-57. [DOI: 10.1007/s13277-014-2812-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/04/2014] [Indexed: 12/25/2022] Open
|
43
|
Abstract
Gastric cancer remains highly prevalent and accounts for a notable proportion of global cancer mortality. This cancer is also associated with poor survival rates. Understanding the genetic basis of gastric cancer will offer insights into its pathogenesis, help identify new biomarkers and novel treatment targets, aid prognostication and could be central to developing individualized treatment strategies in the future. An inherited component contributes to <3% of gastric cancers; the majority of genetic changes associated with gastric cancer are acquired. Over the past few decades, advances in technology and high-throughput analysis have improved understanding of the molecular aspects of the pathogenesis of gastric cancer. These aspects are multifaceted and heterogeneous and represent a wide spectrum of several key genetic influences, such as chromosomal instability, microsatellite instability, changes in microRNA profile, somatic gene mutations or functional single nucleotide polymorphisms. These genetic aspects of the pathogenesis of gastric cancer will be addressed in this Review.
Collapse
Affiliation(s)
- Mairi H McLean
- National Cancer Institute, Laboratory of Molecular Immunoregulation, Cancer &Inflammation Program, 1050 Boyles Street, Frederick, MD 21702-1201, USA
| | - Emad M El-Omar
- Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB51 5ER, UK
| |
Collapse
|
44
|
Yoon JH, Seo HS, Choi SS, Chae HS, Choi WS, Kim O, Ashktorab H, Smoot DT, Nam SW, Lee JY, Park WS. Gastrokine 1 inhibits the carcinogenic potentials of Helicobacter pylori CagA. Carcinogenesis 2014; 35:2619-2629. [PMID: 25239641 PMCID: PMC4303776 DOI: 10.1093/carcin/bgu199] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 08/25/2014] [Accepted: 09/10/2014] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori CagA directly injected by the bacterium into epithelial cells via a type IV secretion system, leads to cellular changes such as morphology, apoptosis, proliferation and cell motility, and stimulates gastric carcinogenesis. We investigated the effects of cytotoxin-associated gene A (CagA) and gastrokine 1 (GKN1) on cell proliferation, apoptosis, reactive oxygen species (ROS) production, epithelial-mesenchymal transition (EMT) and cell migration in CagA- or GKN1-transfected gastric epithelial cells and mucosal tissues from humans and mice infected with H.pylori. On the molecular level, H.pylori CagA induced increased cell proliferation, ROS production, antiapoptotic activity, cell migration and invasion. Moreover, CagA induced activation of NF-κB and PI3K/Akt signaling pathways and EMT-related proteins. In addition, H.pylori CagA reduced GKN1 gene copy number and expression in gastric cells and mucosal tissues of humans and mice. However, GKN1 overexpression successfully suppressed the carcinogenic effects of CagA through binding to CagA. These results suggest that GKN1 might be a target to inhibit the effects from H.pylori CagA.
Collapse
Affiliation(s)
| | - Ho Suk Seo
- Department of General Surgery, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-701, South Korea
| | - Sung Sook Choi
- College of Pharmacy, Sahmyook University, Hwarangro 815, Nowon-gu, Seoul 139-742, South Korea
| | - Hyun Suk Chae
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-701, South Korea
| | | | | | - Hassan Ashktorab
- Department of Medicine, Howard University, Washington, DC 20060, USA
| | - Duane T Smoot
- Department of Internal Medicine, Meharry Medical College, Nashville, TN 37208, USA and
| | - Suk Woo Nam
- Department of Pathology and Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-701, South Korea
| | - Jung Young Lee
- Department of Pathology and Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-701, South Korea
| | - Won Sang Park
- Department of Pathology and Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-701, South Korea
| |
Collapse
|
45
|
Abstract
Inflammatory bowel disease (IBD), which is characterized by chronic or recurrent relapsing gastrointestinal inflammation, includes ulcerative colitis (UC) and Crohn's disease (CD).The precise etiology of IBD remains unclear. In recent years, intestinal mucosal injury is considered the leading cause of IBD, and a large body of evidence suggests that mucins are an important component of the intestinal mucosa barrier and participate in the occurrence and development of IBD. Understanding the relationship between mucins and IBD can provide new avenues for the development of new treatments for this disease.
Collapse
|
46
|
Yoon JH, Choi WS, Kim O, Park WS. The role of gastrokine 1 in gastric cancer. J Gastric Cancer 2014; 14:147-155. [PMID: 25328759 PMCID: PMC4199881 DOI: 10.5230/jgc.2014.14.3.147] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/12/2014] [Accepted: 09/13/2014] [Indexed: 12/18/2022] Open
Abstract
Homeostatic imbalance between cell proliferation and death in gastric mucosal epithelia may lead to gastritis and gastric cancer. Despite abundant gastrokine 1 (GKN1) expression in the normal stomach, the loss of GKN1 expression is frequently detected in gastric mucosa infected with Helicobacter pylori, as well as in intestinal metaplasia and gastric cancer tissues, suggesting that GKN1 plays an important role in gastric mucosal defense, and the gene functions as a gastric tumor suppressor. In the stomach, GKN1 is involved in gastric mucosal inflammation by regulating cytokine production, the nuclear factor-κB signaling pathway, and cyclooxygenase-2 expression. GKN1 also inhibits the carcinogenic potential of H. pylori protein CagA by binding to it, and up-regulates antioxidant enzymes. In addition, GKN1 reduces cell viability, proliferation, and colony formation by inhibiting cell cycle progression and epigenetic modification by down-regulating the expression levels of DNMT1 and EZH2, and DNMT1 activity, and inducing apoptosis through the death receptor-dependent pathway. Furthermore, GKN1 also inhibits gastric cancer cell invasion and metastasis via coordinated regulation of epithelial mesenchymal transition-related protein expression, reactive oxygen species production, and PI3K/Akt signaling pathway activation. Although the modes of action of GKN1 have not been clearly described, recent limited evidence suggests that GKN1 acts as a gastric-specific tumor suppressor. This review aims to discuss, comment, and summarize the recent progress in the understanding of the role of GKN1 in gastric cancer development and progression.
Collapse
Affiliation(s)
- Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Won Suk Choi
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Olga Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Won Sang Park
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
47
|
Dunne C, Dolan B, Clyne M. Factors that mediate colonization of the human stomach by Helicobacter pylori. World J Gastroenterol 2014; 20:5610-24. [PMID: 24914320 PMCID: PMC4024769 DOI: 10.3748/wjg.v20.i19.5610] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/04/2013] [Accepted: 01/19/2014] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori (H. pylori) colonizes the stomach of humans and causes chronic infection. The majority of bacteria live in the mucus layer overlying the gastric epithelial cells and only a small proportion of bacteria are found interacting with the epithelial cells. The bacteria living in the gastric mucus may act as a reservoir of infection for the underlying cells which is essential for the development of disease. Colonization of gastric mucus is likely to be key to the establishment of chronic infection. How H. pylori manages to colonise and survive in the hostile environment of the human stomach and avoid removal by mucus flow and killing by gastric acid is the subject of this review. We also discuss how bacterial and host factors may together go some way to explaining the susceptibility to colonization and the outcome of infection in different individuals. H. pylori infection of the gastric mucosa has become a paradigm for chronic infection. Understanding of why H. pylori is such a successful pathogen may help us understand how other bacterial species colonise mucosal surfaces and cause disease.
Collapse
|
48
|
Shirai K, Okada Y, Cheon DJ, Miyajima M, Behringer RR, Yamanaka O, Saika S. Effects of the loss of conjunctival Muc16 on corneal epithelium and stroma in mice. Invest Ophthalmol Vis Sci 2014; 55:3626-37. [PMID: 24812549 DOI: 10.1167/iovs.13-12955] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE To examine the role of conjunctival Muc16 in the homeostasis of the ocular surface epithelium and stroma using Muc16-null knockout (KO) mice. METHODS We used KO mice (n = 58) and C57/BL6 (WT) mice (n = 58). Histology and immunohistochemistry were employed to analyze the phenotypes in the ocular surface epithelium. The expression of phospho-Stat3, AP-1 components, interleukin 6 (IL-6), and tumor necrosis factor-α (TNFα) in the cornea and conjunctiva was examined. The shape of the nuclei of corneal epithelial cells was examined to evaluate intraepithelial cell differentiation. Epithelial cell proliferation was studied using bromo-deoxyuridine labeling. Finally, the wound healing of a round defect (2-mm diameter) in the corneal epithelium was measured. The keratocyte phenotype and macrophage invasion in the stroma were evaluated after epithelial repair. RESULTS The loss of Muc16 activated Stat3 signal, affected JunB signal, and upregulated the expression of IL-6 in the conjunctiva. Basal-like cells were observed in the suprabasal layer of the corneal epithelium with an increase in proliferation. The loss of Muc16 accelerated the wound healing of the corneal epithelium. The incidence of myofibroblast appearance and macrophage invasion were more marked in KO stroma than in WT stroma after epithelial repair. CONCLUSIONS The loss of Muc16 in the conjunctiva affected the homeostasis of the corneal epithelium and stroma. The mechanism might include the upregulation of the inflammatory signaling cascade (i.e., Stat3 signal, and IL-6 expression in the KO conjunctiva). Current data provides insight into the research of the pathophysiology of dry eye syndrome.
Collapse
Affiliation(s)
- Kumi Shirai
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Dong-Joo Cheon
- Program in Genes and Development, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, United States Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Masayasu Miyajima
- The Laboratory Animal Center, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Richard R Behringer
- Program in Genes and Development, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, United States Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Osamu Yamanaka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, Wakayama, Japan
| |
Collapse
|
49
|
Nath S, Mukherjee P. MUC1: a multifaceted oncoprotein with a key role in cancer progression. Trends Mol Med 2014; 20:332-42. [PMID: 24667139 DOI: 10.1016/j.molmed.2014.02.007] [Citation(s) in RCA: 590] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/16/2014] [Accepted: 02/24/2014] [Indexed: 12/15/2022]
Abstract
The transmembrane glycoprotein Mucin 1 (MUC1) is aberrantly glycosylated and overexpressed in a variety of epithelial cancers, and plays a crucial role in progression of the disease. Tumor-associated MUC1 differs from the MUC1 expressed in normal cells with regard to its biochemical features, cellular distribution, and function. In cancer cells, MUC1 participates in intracellular signal transduction pathways and regulates the expression of its target genes at both the transcriptional and post-transcriptional levels. This review highlights the structural and functional differences that exist between normal and tumor-associated MUC1. We also discuss the recent advances made in the use of MUC1 as a biomarker and therapeutic target for cancer.
Collapse
Affiliation(s)
- Sritama Nath
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Pinku Mukherjee
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC, USA.
| |
Collapse
|
50
|
Guang W, Czinn SJ, Blanchard TG, Kim KC, Lillehoj EP. Genetic regulation of MUC1 expression by Helicobacter pylori in gastric cancer cells. Biochem Biophys Res Commun 2014; 445:145-50. [PMID: 24491543 DOI: 10.1016/j.bbrc.2014.01.142] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 01/26/2014] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori infection of the stomach is associated with the development of gastritis, peptic ulcers, and gastric adenocarcinomas, but the mechanisms are unknown. MUC1 is aberrantly overexpressed by more than 50% of stomach cancers, but its role in carcinogenesis remains to be defined. The current studies were undertaken to identify the genetic mechanisms regulating H. pylori-dependent MUC1 expression by gastric epithelial cells. Treatment of AGS cells with H. pylori increased MUC1 mRNA and protein levels, and augmented MUC1 gene promoter activity, compared with untreated cells. H. pylori increased binding of STAT3 and MUC1 itself to the MUC1 gene promoter within a region containing a STAT3 binding site, and decreased CpG methylation of the MUC1 promoter proximal to the STAT3 binding site, compared with untreated cells. These results suggest that H. pylori upregulates MUC1 expression in gastric cancer cells through STAT3 and CpG hypomethylation.
Collapse
Affiliation(s)
- Wei Guang
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Steven J Czinn
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Thomas G Blanchard
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - K Chul Kim
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, United States
| | - Erik P Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|