1
|
Pinho AS, Pereira R, Pereira M, Rai A, Ferreira L, Martins MCL, Parreira P. Cholesterol Functionalized Nanoparticles Are Effective against Helicobacter pylori, the Gastric Bug: A Proof-of-Concept Study. Adv Healthc Mater 2025:e2404065. [PMID: 39910897 DOI: 10.1002/adhm.202404065] [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: 10/18/2024] [Revised: 01/20/2025] [Indexed: 02/07/2025]
Abstract
Helicobacter pylori chronic infection is the highest risk factor for the development of gastric cancer, being this Gram-negative bacterium classified as carcinogenic. The mounting resistance of H. pylori to antibiotics calls for innovative therapeutic strategies. Here, the proof-of-concept studies that support the development of a "trojan horse" therapeutic strategy based on cholesterol-grafted nanoparticles (Chol-NP) to counteract H. pylori infection are depicted. The bacterium ability to specifically recognize and bind to surface grafted cholesterol is demonstrated by its adhesion to cholesterol(Chol)-functionalized self-assembled monolayers (SAMs) on gold substrates (2D Chol-SAMs) in a concentration dependent manner, with optimal Chol-SAMs prepared with 25% Chol-polyethylene glycol (PEG)-thiol in solution (75% tetra(ethylene glycol)-thiol). These results further show that cholesterol functionalized gold nanoparticles (3D Chol-SAMs, Chol-NP) eradicate H. pylori at a minimum bactericidal concentration of 125 µg mL-1. Chol-NP kill H. pylori through internalization and membrane rupture, as observed by transmission electron microscopy (TEM). Chol-NP are cytocompatible (human gastric adenocarcinoma (AGS) cell line), non-hemolytic and innocuous to bacteria representative of the gut microbiota (Escherichia coli and Lactobacillus acidophilus). This study supports the further development of cholesterol functionalized biomaterials as an advanced and targeted treatment for H. pylori infection.
Collapse
Affiliation(s)
- Ana Sofia Pinho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Renato Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
- FEUP - Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, Porto, 4200-465, Portugal
| | - Mariana Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
| | - Akhilesh Rai
- CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Rua Larga, 3004-504, Portugal
| | - Lino Ferreira
- CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Rua Larga, 3004-504, Portugal
- FMUC - Faculdade de Medicina, Universidade de Coimbra, Azinhaga de Santa Comba (Celas), 3000-548, Portugal
- CIBB- Centre for Innovative Biomedicine and Biotechnology Associate Laboratory, Universidade de Coimbra, Rua Larga, 3004-504, Portugal
| | - Maria Cristina Lopes Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Paula Parreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, Porto, 4200-135, Portugal
| |
Collapse
|
2
|
Song M, Chen S, Lin W, Zhu K. Targeting bacterial phospholipids and their synthesis pathways for antibiotic discovery. Prog Lipid Res 2024; 96:101307. [PMID: 39566858 DOI: 10.1016/j.plipres.2024.101307] [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: 08/15/2024] [Revised: 10/31/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Bacterial infections in humans and animals caused by multidrug-resistant (MDR) pathogens pose a serious threat to public health. New antibacterial targets are extremely urgent to solve the dilemma of cross-resistance. Phospholipids are critical components in bacterial envelopes and involve diverse crucial processes to maintain homeostasis and modulate metabolism. Targeting phospholipids and their synthesis pathways has been largely overlooked because conventional membrane-targeted substances are non-specific with cytotoxicity. In this review, we first introduce the structure and physiological function of phospholipids in bacteria. Subsequently, we describe the chemical diversity of novel ligands targeting phospholipids, structure-activity relationships (SAR), modes of action (MOA), and pharmacological effects. Finally, we prospect the advantage of bacterial phospholipids as promising antibacterial targets. In conclusion, these findings will shed light on discovering and developing new antibacterial drugs to combat MDR bacteria-associated infections.
Collapse
Affiliation(s)
- Meirong Song
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Shang Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Wenhan Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Kui Zhu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
3
|
Chen M, Wu Z, Zou Y, Peng C, Hao Y, Zhu Z, Shi X, Su B, Ou L, Lai Y, Jia J, Xun M, Li H, Zhu W, Feng Z, Yao M. Phellodendron chinense C.K.Schneid: An in vitro study on its anti-Helicobacter pylori effect. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118396. [PMID: 38823658 DOI: 10.1016/j.jep.2024.118396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/15/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phellodendron chinense C.K.Schneid(P. chinense Schneid) is known in TCM as Huang Bo, is traditionally used to support gastrointestinal function and alleviate stomach-related ailments, including gastric ulcer bleeding and symptoms of gastroesophageal reflux disease. Helicobacter pylori (H. pylori) is classified by the WHO as a Group 1 carcinogen. However, the specific activity and mechanism of action of P. chinense Schneid against H. pylori infection remain unclear. It has been noted that Huangjiu processing may alter the bitter and cold properties of P. chinense Schneid, but its effect on antimicrobial activity requires further investigation. Additionally, it remains uncertain whether berberine is the sole antimicrobial active component of P. chinense Schneid. AIM OF STUDY This study aims to elucidate the anti-H. pylori infection activity of P. chinense Schneid, along with its mechanism of action and key antimicrobial active components. MATERIALS AND METHODS Phytochemical analysis was carried out by UPLC-MS/MS. HPLC was employed to quantify the berberine content of the extracts. Antimicrobial activity was assessed using the micro broth dilution method. Morphology was observed using SEM. The impact on urease activity was analyzed through in vitro urease enzyme kinetics. RT-qPCR was employed to detect the expression of virulence genes, including adhesin, flagellum, urease, and cytotoxin-related genes. The adhesion effect was evaluated by immunofluorescence staining and agar culture. RESULTS P. chinense Schneid exhibited strong antimicrobial activity against both antibiotic-sensitive and resistant H. pylori strains, with MIC ranging from 40 to 160 μg/mL. Combination with amoxicillin, metronidazole, levofloxacin, and clarithromycin did not result in antagonistic effects. P. chinense Schneid induced alterations in bacterial morphology and structure, downregulated the expression of various virulence genes, and inhibited urease enzyme activity. In co-infection systems, P. chinense Schneid significantly attenuated H. pylori adhesion and urease relative content, thereby mitigating cellular damage caused by infection. Huangjiu processing enhanced the anti-H. pylori activity of P. chinense Schneid. Besides berberine, P. chinense Schneid contained seven other components with anti-H. pylori activity, with palmatine exhibiting the strongest activity, followed by jatrorrhizine. CONCLUSIONS This study sheds light on the potential therapeutic mechanisms of P. chinense Schneid against H. pylori infection, demonstrating its capacity to disrupt bacterial structure, inhibit urease activity, suppress virulence gene transcription, inhibit adhesion, and protect host cells. The anti-H. pylori activity of P. chinense Schneid was potentiated by Huangjiu processing, and additional components beyond berberine were identified as possessing strong anti-H. pylori activity. Notably, jatrorrhizine, a core component of P. chinense Schneid, exhibited significant anti-H. pylori activity, marking a groundbreaking discovery.
Collapse
Affiliation(s)
- Meiyun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Ziyao Wu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Yuanjing Zou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Chang Peng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yajie Hao
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Zhixiang Zhu
- School of Medicine and Pharmacy (Qingdao), Ocean University of China, Qingdao, 266003, China.
| | - Xiaoyan Shi
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Bingmei Su
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Ling Ou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yuqian Lai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Junwei Jia
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Mingjin Xun
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Hui Li
- Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Weixing Zhu
- Qingyuan Hospital of Traditional Chinese Medicine, Qingyuan, 511500, China.
| | - Zhong Feng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China; International Pharmaceutical Engineering Lab of Shandong Province, Feixian, 273400, China; Lunan Pharmaceutical Group Co., Ltd, Linyi, 276000, Shandong, China.
| | - Meicun Yao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
4
|
Chen PH, Tsai CL, Hsieh YW, Cho DY, Tsai FJ, Lin CL, Liao HY. Antihyperlipidemic drugs mitigate the elevated incidence of peptic ulcer disease caused by hyperlipidemia: A cohort study. J Chin Med Assoc 2024; 87:961-968. [PMID: 39118217 DOI: 10.1097/jcma.0000000000001145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Several risk factors for peptic ulcer disease (PUD) have been identified; however, the recurrence rate of PUD remains high even with standard ulcer treatments. High cholesterol levels have been proposed as a risk factor for PUD, but clinical evidence remains limited. Therefore, this database study investigated whether hyperlipidemia increases PUD risk and whether antihyperlipidemic drugs reduce this risk. METHODS A long-term cohort design was adopted, and Taiwan's National Health Insurance Research Database was used to enroll patients diagnosed with hyperlipidemia between 2000 and 2016. Patients without hyperlipidemia were randomly matched based on variables such as age and gender to establish a comparison cohort at a 1:1 ratio. Another cohort study was conducted to determine whether antihyperlipidemic drugs or red yeast rice prescriptions can reduce the incidence of PUD in patients with hyperlipidemia. RESULTS The overall incidence of PUD was 1.48 times higher in the hyperlipidemia cohort (203,235 patients) than in the nonhyperlipidemia cohort (adjusted hazard ratio, 1.48; 95% CI, 1.46-1.50; p < 0.001). Among the patients with hyperlipidemia, those who used antihyperlipidemic drugs with or without red yeast rice prescriptions exhibited a lower risk of developing PUD relative to those who did not use them; the adjusted hazard ratios were 0.33 (95% CI, 0.21-0.52) and 0.81 (95% CI, 0.78-0.84), respectively. When the cumulative exposure to antihyperlipidemic drugs and red yeast rice prescriptions increased, the risk of developing PUD showed a decreasing trend, which was statistically significant for antihyperlipidemic drugs but not for red yeast rice. CONCLUSION Hyperlipidemia is associated with a higher risk of PUD, which can be reduced through the administration of antihyperlipidemic drugs with or without red yeast rice prescriptions.
Collapse
Affiliation(s)
- Pei-Hsien Chen
- Department of Chinese Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
| | - Chiu-Lin Tsai
- Department of Chinese Medicine Pharmacy, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
| | - Yow-Wen Hsieh
- Department of Pharmacy, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
- Graduate Institute of Pharmacy, China Medical University, Taichung, Taiwan, ROC
| | - Der-Yang Cho
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
- Department of Neurosurgery, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan, ROC
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
- Division of Medical Genetics, China Medical University Children's Hospital, China Medical University, Taichung, Taiwan, ROC
- Department of Medical Laboratory Science & Biotechnology, Asia University, Taichung, Taiwan, ROC
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, China Medical University, Taiwan, ROC
| | - Hsien-Yin Liao
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan, ROC
- Department of Acupuncture, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
| |
Collapse
|
5
|
Lin CM, Meng Q, Li YJ, Zhang SX, Luo QX, Dai ZY. Causal associations between intermediate very-low-density lipoprotein cholesterol-to-total lipids ratio and peptic ulcer: A bidirectional Mendelian randomization study. World J Clin Cases 2024; 12:5729-5738. [PMID: 39247748 PMCID: PMC11263067 DOI: 10.12998/wjcc.v12.i25.5729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Previous epidemiologic investigations have consistently demonstrated a strong association between the ratio of cholesterol to total lipids in medium very-low-density lipoprotein (VLDL) and the occurrence of peptic ulcers (PU). However, the precise causal relationship between these factors remains ambiguous. Consequently, this study aims to elucidate the potential correlation between the ratio of cholesterol to total lipids in medium VLDL and the incidence of peptic ulcer. AIM To investigate the ratio of cholesterol to total lipids in medium very-low-density lipoprotein (VLDL) association with PU via genetic methods, guiding future clinical research. METHODS Genome-wide association study (GWAS) datasets for the ratio of cholesterol to total lipids in intermediate VLDL and peptic ulcer were retrieved from the IEU OpenGWAS project (https://gwas.mrcieu.ac.uk). For the forward Mendelian randomization (MR) analysis, 72 single nucleotide polymorphisms (SNPs) were identified as instrumental variables. These SNPs were selected based on their association with the ratio of cholesterol to total lipids in intermediate VLDL, with peptic ulcer as the outcome variable. Conversely, for the inverse MR analysis, no SNPs were identified with peptic ulcer as the exposure variable and the ratio of cholesterol to total lipids in intermediate VLDL as the outcome. All MR analyses utilized inverse variance weighted (IVW) as the primary analytical method. Additionally, weighted median and MR-Egger methods were employed as supplementary analytical approaches to assess causal effects. Egger regression was used as a supplementary method to evaluate potential directional pleiotropy. Heterogeneity and multiplicity tests were conducted using the leave-one-out method to evaluate result stability and mitigate biases associated with multiple testing. RESULTS The genetically predicted ratio of cholesterol to total lipids in medium VLDL was significantly associated with an elevated risk of peptic ulcer (IVW: OR = 2.557, 95%CI = 1.274-5.132, P = 0.008). However, no causal association of peptic ulcer with the ratio of cholesterol to total lipids in medium VLDL was observed in the inverse Mendelian randomization analysis. CONCLUSION In conclusion, our study reveals a significant association between the ratio of cholesterol to total lipids in medium VLDL and an elevated risk of peptic ulcers. However, further validation through laboratory investigations and larger-scale studies is warranted to strengthen the evidence and confirm the causal relationship between these factors.
Collapse
Affiliation(s)
- Chun-Mei Lin
- Postgraduate Student, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Qian Meng
- Postgraduate Student, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Ying-Jun Li
- Postgraduate Student, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Shuang-Xi Zhang
- Department of Gastroenterology, Guangzhou University of Chinese Medicine Shunde Hospital, Foshan 528300, Guangdong Province, China
| | - Qiong-Xi Luo
- Postgraduate Student, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Zhen-Yu Dai
- Postgraduate Student, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| |
Collapse
|
6
|
Fei X, Li N, Xu X, Zhu Y. Macrophage biology in the pathogenesis of Helicobacter pylori infection. Crit Rev Microbiol 2024:1-18. [PMID: 39086061 DOI: 10.1080/1040841x.2024.2366944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Infection with H. pylori induces chronic gastric inflammation, progressing to peptic ulcer and stomach adenocarcinoma. Macrophages function as innate immune cells and play a vital role in host immune defense against bacterial infection. However, the distinctive mechanism by which H. pylori evades phagocytosis allows it to colonize the stomach and further aggravate gastric preneoplastic pathology. H. pylori exacerbates gastric inflammation by promoting oxidative stress, resisting macrophage phagocytosis, and inducing M1 macrophage polarization. M2 macrophages facilitate the proliferation, invasion, and migration of gastric cancer cells. Various molecular mechanisms governing macrophage function in the pathogenesis of H. pylori infection have been identified. In this review, we summarize recent findings of macrophage interactions with H. pylori infection, with an emphasis on the regulatory mechanisms that determine the clinical outcome of bacterial infection.
Collapse
Affiliation(s)
- Xiao Fei
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nianshuang Li
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xinbo Xu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
7
|
Borsato G, Carnio F, Lunardon S, Moletta M, Pavan G, Terrin F, Scarso A, Plotegher N, Fabris F. A β-Glucosyl Sterol Probe for in situ Fluorescent Labelling in Neuronal Cells to Investigate Neurodegenerative Diseases. Chemistry 2024; 30:e202400778. [PMID: 38770991 DOI: 10.1002/chem.202400778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 05/22/2024]
Abstract
A β-glucosyl sterol probe bearing a terminal alkyne moiety for fluorescent tagging enables the investigation of the neuronal and intracellular localization of this class of compounds involved in neurodegenerative diseases. The compound showed localization in the neuronal cells, with marked differences in the uptake and metabolism leading to enhanced persistence with respect to the un-glycosylated sterol analogue. In addition, a different impact was observed towards lysosomes, with the simple sterol probe showing the enlargement of the lysosome structures, while the β-glucosyl sterol was less capable to alter the morphology of this specific organelle.
Collapse
Affiliation(s)
- Giuseppe Borsato
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Francesco Carnio
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Sara Lunardon
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Mattia Moletta
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Giulio Pavan
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Francesca Terrin
- Dipartimento di Biologia, Università degli Studi di Padova, viale G. Colombo 3, 35131, Padova, Italy
| | - Alessandro Scarso
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| | - Nicoletta Plotegher
- Dipartimento di Biologia, Università degli Studi di Padova, viale G. Colombo 3, 35131, Padova, Italy
| | - Fabrizio Fabris
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, via Torino 155, 30172, Mestre Venezia, Italy
| |
Collapse
|
8
|
Dore MP, Pes GM. Trained Immunity and Trained Tolerance: The Case of Helicobacter pylori Infection. Int J Mol Sci 2024; 25:5856. [PMID: 38892046 PMCID: PMC11172748 DOI: 10.3390/ijms25115856] [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: 04/24/2024] [Revised: 05/16/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Trained immunity is a concept in immunology in which innate immune cells, such as monocytes and macrophages, exhibit enhanced responsiveness and memory-like characteristics following initial contact with a pathogenic stimulus that may promote a more effective immune defense following subsequent contact with the same pathogen. Helicobacter pylori, a bacterium that colonizes the stomach lining, is etiologically associated with various gastrointestinal diseases, including gastritis, peptic ulcer, gastric adenocarcinoma, MALT lymphoma, and extra gastric disorders. It has been demonstrated that repeated exposure to H. pylori can induce trained immunity in the innate immune cells of the gastric mucosa, which become more responsive and better able to respond to subsequent H. pylori infections. However, interactions between H. pylori and trained immunity are intricate and produce both beneficial and detrimental effects. H. pylori infection is characterized histologically as the presence of both an acute and chronic inflammatory response called acute-on-chronic inflammation, or gastritis. The clinical outcomes of ongoing inflammation include intestinal metaplasia, gastric atrophy, and dysplasia. These same mechanisms may also reduce immunotolerance and trigger autoimmune pathologies in the host. This review focuses on the relationship between trained immunity and H. pylori and underscores the dynamic interplay between the immune system and the pathogen in the context of gastric colonization and inflammation.
Collapse
Affiliation(s)
- Maria Pina Dore
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Clinica Medica, Viale San Pietro 8, 07100 Sassari, Italy;
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza Blvd, Houston, TX 77030, USA
| | - Giovanni Mario Pes
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Clinica Medica, Viale San Pietro 8, 07100 Sassari, Italy;
| |
Collapse
|
9
|
Ong LL, Jan HM, Le HHT, Yang TC, Kuo CY, Feng AF, Mong KKT, Lin CH. Membrane lipid remodeling eradicates Helicobacter pylori by manipulating the cholesteryl 6'-acylglucoside biosynthesis. J Biomed Sci 2024; 31:44. [PMID: 38685037 PMCID: PMC11057186 DOI: 10.1186/s12929-024-01031-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Helicobacter pylori, the main cause of various gastric diseases, infects approximately half of the human population. This pathogen is auxotrophic for cholesterol which it converts to various cholesteryl α-glucoside derivatives, including cholesteryl 6'-acyl α-glucoside (CAG). Since the related biosynthetic enzymes can be translocated to the host cells, the acyl chain of CAG likely comes from its precursor phosphatidylethanolamine (PE) in the host membranes. This work aims at examining how the acyl chain of CAG and PE inhibits the membrane functions, especially bacterial adhesion. METHODS Eleven CAGs that differ in acyl chains were used to study the membrane properties of human gastric adenocarcinoma cells (AGS cells), including lipid rafts clustering (monitored by immunofluorescence with confocal microscopy) and lateral membrane fluidity (by the fluorescence recovery after photobleaching). Cell-based and mouse models were employed to study the degree of bacterial adhesion, the analyses of which were conducted by using flow cytometry and immunofluorescence staining, respectively. The lipidomes of H. pylori, AGS cells and H. pylori-AGS co-cultures were analyzed by Ultraperformance Liquid Chromatography-Tandem Mass Spectroscopy (UPLC-MS/MS) to examine the effect of PE(10:0)2, PE(18:0)2, PE(18:3)2, or PE(22:6)2 treatments. RESULTS CAG10:0, CAG18:3 and CAG22:6 were found to cause the most adverse effect on the bacterial adhesion. Further LC-MS analysis indicated that the treatment of PE(10:0)2 resulted in dual effects to inhibit the bacterial adhesion, including the generation of CAG10:0 and significant changes in the membrane compositions. The initial (1 h) lipidome changes involved in the incorporation of 10:0 acyl chains into dihydro- and phytosphingosine derivatives and ceramides. In contrast, after 16 h, glycerophospholipids displayed obvious increase in their very long chain fatty acids, monounsaturated and polyunsaturated fatty acids that are considered to enhance membrane fluidity. CONCLUSIONS The PE(10:0)2 treatment significantly reduced bacterial adhesion in both AGS cells and mouse models. Our approach of membrane remodeling has thus shown great promise as a new anti-H. pylori therapy.
Collapse
Affiliation(s)
- Lih-Lih Ong
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, 1001, University Road, Eastern District, Hsinchu, 300093, Taiwan
- Institute of Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Hau-Ming Jan
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Hong-Hanh Thi Le
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Tsai-Chen Yang
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Chou-Yu Kuo
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Ai-Feng Feng
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, 1001, University Road, Eastern District, Hsinchu, 300093, Taiwan
| | - Kwok-Kong Tony Mong
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, 1001, University Road, Eastern District, Hsinchu, 300093, Taiwan.
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.
- Department of Chemistry and Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
10
|
Rosli NA, Al-Maleki AR, Loke MF, Tay ST, Rofiee MS, Teh LK, Salleh MZ, Vadivelu J. Exposure of Helicobacter pylori to clarithromycin in vitro resulting in the development of resistance and triggers metabolic reprogramming associated with virulence and pathogenicity. PLoS One 2024; 19:e0298434. [PMID: 38446753 PMCID: PMC10917248 DOI: 10.1371/journal.pone.0298434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/23/2024] [Indexed: 03/08/2024] Open
Abstract
In H. pylori infection, antibiotic-resistance is one of the most common causes of treatment failure. Bacterial metabolic activities, such as energy production, bacterial growth, cell wall construction, and cell-cell communication, all play important roles in antimicrobial resistance mechanisms. Identification of microbial metabolites may result in the discovery of novel antimicrobial therapeutic targets and treatments. The purpose of this work is to assess H. pylori metabolomic reprogramming in order to reveal the underlying mechanisms associated with the development of clarithromycin resistance. Previously, four H. pylori isolates were induced to become resistant to clarithromycin in vitro by incrementally increasing the concentrations of clarithromycin. Bacterial metabolites were extracted using the Bligh and Dyer technique and analyzed using metabolomic fingerprinting based on Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry (LC-Q-ToF-MS). The data was processed and analyzed using the MassHunter Qualitative Analysis and Mass Profiler Professional software. In parental sensitivity (S), breakpoint isolates (B), and induced resistance isolates (R) H. pylori isolates, 982 metabolites were found. Furthermore, based on accurate mass, isotope ratios, abundances, and spacing, 292 metabolites matched the metabolites in the Agilent METLIN precise Mass-Personal Metabolite Database and Library (AM-PCDL). Several metabolites associated with bacterial virulence, pathogenicity, survival, and proliferation (L-leucine, Pyridoxone [Vitamine B6], D-Mannitol, Sphingolipids, Indoleacrylic acid, Dulcitol, and D-Proline) were found to be elevated in generated resistant H. pylori isolates when compared to parental sensitive isolates. The elevated metabolites could be part of antibiotics resistance mechanisms. Understanding the fundamental metabolome changes in the course of progressing from clarithromycin-sensitive to breakpoint to resistant in H. pylori clinical isolates may be a promising strategy for discovering novel alternatives therapeutic targets.
Collapse
Affiliation(s)
- Naim Asyraf Rosli
- Faculty of Medicine, Department of Medical Microbiology, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Anis Rageh Al-Maleki
- Faculty of Medicine, Department of Medical Microbiology, Universiti Malaya, Kuala Lumpur, Malaysia
- Faculty of Medicine and Health Sciences, Department of Medical Microbiology, Sana’a University, Sana’a, Yemen
| | - Mun Fai Loke
- Camtech Biomedical Pte Ltd, Singapore, Singapore
| | - Sun Tee Tay
- Faculty of Medicine, Department of Medical Microbiology, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Mohd Salleh Rofiee
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA, Selangor, Malaysia
| | - Lay Kek Teh
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA, Selangor, Malaysia
| | - Mohd Zaki Salleh
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA, Selangor, Malaysia
| | - Jamuna Vadivelu
- Faculty of Medicine, Medical Education Research and Development Unit (MERDU), Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
11
|
Sit WY, Cheng ML, Chen TJ, Chen CJ, Chen BN, Huang DJ, Chen PL, Chen YC, Lo CJ, Wu DC, Hsieh WC, Chang CT, Chen RH, Wang WC. Helicobacter pylori PldA modulates TNFR1-mediated p38 signaling pathways to regulate macrophage responses for its survival. Gut Microbes 2024; 16:2409924. [PMID: 39369445 PMCID: PMC11457642 DOI: 10.1080/19490976.2024.2409924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/16/2024] [Accepted: 09/11/2024] [Indexed: 10/08/2024] Open
Abstract
Helicobacter pylori, a dominant member of the gastric microbiota was associated with various gastrointestinal diseases and presents a significant challenge due to increasing antibiotic resistance. This study identifies H. pylori's phospholipase A (PldA) as a critical factor in modulating host macrophage responses, facilitating H. pylori 's evasion of the immune system and persistence. PldA alters membrane lipids through reversible acylation and deacylation, affecting their structure and function. We found that PldA incorporates lysophosphatidylethanolamine into macrophage membranes, disrupting their bilayer structure and impairing TNFR1-mediated p38-MK2 signaling. This disruption results in reduced macrophage autophagy and elevated RIP1-dependent apoptosis, thereby enhancing H. pylori survival, a mechanism also observed in multidrug-resistant strains. Pharmacological inhibition of PldA significantly decreases H. pylori viability and increases macrophage survival. In vivo studies corroborate PldA's essential role in H. pylori persistence and immune cell recruitment. Our findings position PldA as a pivotal element in H. pylori pathogenesis through TNFR1-mediated membrane modulation, offering a promising therapeutic target to counteract bacterial resistance.
Collapse
Affiliation(s)
- Wei Yang Sit
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Mei-Ling Cheng
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Tsan-Jan Chen
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Chia-Jo Chen
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Bo-Nian Chen
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Ding-Jun Huang
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Pei-Lien Chen
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Yun-Ching Chen
- Institute of Biomedical Engineering, National Tsing-Hua University, Hsinchu, Taiwan, ROC
| | - Chi-Jen Lo
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
| | - Wan-Chen Hsieh
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Chung-Ting Chang
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Ruey-Hwa Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan, ROC
| | - Wen-Ching Wang
- Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| |
Collapse
|
12
|
Ascari A, Frölich S, Zang M, Tran ENH, Wilson DW, Morona R, Eijkelkamp BA. Shigella flexneri remodeling and consumption of host lipids during infection. J Bacteriol 2023; 205:e0032023. [PMID: 37991380 PMCID: PMC10729657 DOI: 10.1128/jb.00320-23] [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: 09/26/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
IMPORTANCE Bacterial pathogens have vastly distinct sites that they inhabit during infection. This requires adaptation due to changes in nutrient availability and antimicrobial stress. The bacterial surface is a primary barrier, and here, we show that the bacterial pathogen Shigella flexneri increases its surface decorations when it transitions to an intracellular lifestyle. We also observed changes in bacterial and host cell fatty acid homeostasis. Specifically, intracellular S. flexneri increased the expression of their fatty acid degradation pathway, while the host cell lipid pool was significantly depleted. Importantly, bacterial proliferation could be inhibited by fatty acid supplementation of host cells, thereby providing novel insights into the possible link between human malnutrition and susceptibility to S. flexneri.
Collapse
Affiliation(s)
- Alice Ascari
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| | - Sonja Frölich
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, Australia
| | - Maoge Zang
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| | - Elizabeth N. H. Tran
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| | - Danny W. Wilson
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, Australia
| | - Renato Morona
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| | - Bart A. Eijkelkamp
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| |
Collapse
|
13
|
Wanibuchi K, Hosoda K, Amgalanbaatar A, Ihara M, Takezawa M, Sakai Y, Masui H, Shoji M, Hayashi S, Shimomura H. Aspects for development of novel antibacterial medicines using a vitamin D 3 decomposition product in Helicobacter pylori infection. J Antibiot (Tokyo) 2023; 76:665-672. [PMID: 37658133 DOI: 10.1038/s41429-023-00651-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/05/2023] [Accepted: 08/10/2023] [Indexed: 09/03/2023]
Abstract
A previous study by our group demonstrated that a vitamin D3 decomposition product (VDP1) acts as the selective bactericidal substance on Helicobacter pylori. VDP1 is an indene compound modified with a carbonyl and an alkyl. The alkyl of VDP1 turned out to be a mandatory structure to exert effective bactericidal action on H. pylori. Meanwhile, it still remains to be clarified as to how influence the alteration of the carbonyl in VDP1 has on the anti-H. pylori activity. In this study, we synthesized novel VDP1 derivatives that replaced the carbonyl of VDP1 by various functional groups and investigated the antibacterial action of the VDP1 derivatives on H. pylori. VDP1 derivatives retaining either a hydroxy (VD3-1) or an acetic ester (VD3-3) exhibited more effective bactericidal action to H. pylori than VDP1. The replacement of the carbonyl of VDP1 by either an allyl acetate (VD3-2) or an acrylic acid (VD3-5) provided almost no change to the anti-H. pylori activity. Apart from this, an isomer of VDP1 (VD3-4) slightly improved anti-H. pylori activity of VDP1. Meanwhile, the replacement of the carbonyl of VDP1 by a methyl acrylate (VD3-6) attenuated the anti-H. pylori activity. As with VDP1, its derivatives also were suggested to exert the anti-H. pylori action through the interaction with myristic acid side chains of dimyristoyl-phosphatidylethanolamine, a characteristic membrane lipid constituent of this pathogen. These results indicate that it is capable of developing specific antibacterial medicines for H. pylori targeting the biomembranal dimyristoyl-phosphatidylethanolamine using VDP1 as the fundamental structure.
Collapse
Affiliation(s)
- Kiyofumi Wanibuchi
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Kouichi Hosoda
- Nikon Cell Innovation Co., Ltd., 2-4-10, Shinsuna, Koto-ku, Tokyo, 136-0075, Japan
| | - Avarzed Amgalanbaatar
- Department of Graduate Education, Graduate School, Mongolian National University of Medical Sciences, 14210, Zoing street, Sukhbaatar District, Ulaanbaatar, 14210, Mongolia
| | - Masato Ihara
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Motoki Takezawa
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Yuki Sakai
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Hisashi Masui
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Mitsuru Shoji
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan
| | - Shunji Hayashi
- Department of Microbiology, School of Medicine, Kitasato University, 1-15-1, Kitasato, Minami-ku, Sagamihara-shi, Kanagawa, 252-0374, Japan
| | - Hirofumi Shimomura
- Public Health Center of Uki, Kumamoto Prefecture Office, 400-1, Kugu, Matsubase-machi, Uki-shi, Kumamoto, 869-0532, Japan.
| |
Collapse
|
14
|
Prado LG, Camara NOS, Barbosa AS. Cell lipid biology in infections: an overview. Front Cell Infect Microbiol 2023; 13:1148383. [PMID: 37868347 PMCID: PMC10587689 DOI: 10.3389/fcimb.2023.1148383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Lipids are a big family of molecules with a vast number of functions in the cell membranes, within the cytoplasm, and extracellularly. Lipid droplets (LDs) are the most common storage organelles and are present in almost every tissue type in the body. They also have structural functions serving as building blocks of cellular membranes and may be precursors of other molecules such as hormones, and lipoproteins, and as messengers in signal transduction. Fatty acids (FAs), such as sterol esters and triacylglycerols, are stored in LDs and are used in β-oxidation as fuel for tricarboxylic acid cycle (TCA) and adenosine triphosphate (ATP) generation. FA uptake and entrance in the cytoplasm are mediated by membrane receptors. After a cytoplasmic round of α- and β-oxidation, FAs are guided into the mitochondrial matrix by the L-carnitine shuttle system, where they are fully metabolized, and enter the TCA cycle. Pathogen infections may lead to impaired lipid metabolism, usage of membrane phospholipids, and LD accumulation in the cytoplasm of infected cells. Otherwise, bacterial pathogens may use lipid metabolism as a carbon source, thus altering the reactions and leading to cellular and organelles malfunctioning. This review aims to describe cellular lipid metabolism and alterations that occur upon infections.
Collapse
Affiliation(s)
- Luan Gavião Prado
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Camara
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
15
|
Wizenty J, Koop PH, Clusmann J, Tacke F, Trautwein C, Schneider KM, Sigal M, Schneider CV. Association of Helicobacter pylori Positivity With Risk of Disease and Mortality. Clin Transl Gastroenterol 2023; 14:e00610. [PMID: 37367296 PMCID: PMC10522101 DOI: 10.14309/ctg.0000000000000610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/28/2023] Open
Abstract
INTRODUCTION Helicobacter pylori colonizes the human stomach. Infection causes chronic gastritis and increases the risk of gastroduodenal ulcer and gastric cancer. Its chronic colonization in the stomach triggers aberrant epithelial and inflammatory signals that are also associated with systemic alterations. METHODS Using a PheWAS analysis in more than 8,000 participants in the community-based UK Biobank, we explored the association of H. pylori positivity with gastric and extragastric disease and mortality in a European country. RESULTS Along with well-established gastric diseases, we dominantly found overrepresented cardiovascular, respiratory, and metabolic disorders. Using multivariate analysis, the overall mortality of H. pylori -positive participants was not altered, while the respiratory and Coronovirus 2019-associated mortality increased. Lipidomic analysis for H. pylori -positive participants revealed a dyslipidemic profile with reduced high-density lipoprotein cholesterol and omega-3 fatty acids, which may represent a causative link between infection, systemic inflammation, and disease. DISCUSSION Our study of H. pylori positivity demonstrates that it plays an organ- and disease entity-specific role in the development of human disease and highlights the importance of further research into the systemic effects of H. pylori infection.
Collapse
Affiliation(s)
- Jonas Wizenty
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Paul-Henry Koop
- Department for Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Jan Clusmann
- Department for Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Trautwein
- Department for Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Kai Markus Schneider
- Department for Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Sigal
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carolin V. Schneider
- Department for Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
16
|
Innate immune activation and modulatory factors of Helicobacter pylori towards phagocytic and nonphagocytic cells. Curr Opin Immunol 2023; 82:102301. [PMID: 36933362 DOI: 10.1016/j.coi.2023.102301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023]
Abstract
Helicobacter pylori is an intriguing obligate host-associated human pathogen with a specific host interaction biology, which has been shaped by thousands of years of host-pathogen coevolution. Molecular mechanisms of interaction of H. pylori with the local immune cells in the human system are less well defined than epithelial cell interactions, although various myeloid cells, including neutrophils and other phagocytes, are locally present or attracted to the sites of infection and interact with H. pylori. We have recently addressed the question of novel bacterial innate immune stimuli, including bacterial cell envelope metabolites, that can activate and modulate cell responses via the H. pylori Cag type IV secretion system. This review article gives an overview of what is currently known about the interaction modes and mechanisms of H. pylori with diverse human cell types, with a focus on bacterial metabolites and cells of the myeloid lineage including phagocytic and antigen-presenting cells.
Collapse
|
17
|
Hosoda K, Wanibuchi K, Amgalanbaatar A, Shoji M, Hayashi S, Shimomura H. A novel role of catalase in cholesterol uptake of Helicobacter pylori. Steroids 2023; 191:109158. [PMID: 36574870 DOI: 10.1016/j.steroids.2022.109158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 12/18/2022] [Indexed: 12/25/2022]
Abstract
Helicobacter pylori infection is known to be a significant risk factor for the development of gastric cancers in humans. This pathogen exhibits unique biological characteristics in membrane lipid composition. Specifically, H. pylori incorporates exogenous cholesterol into biomembranes and uses cholesterol as the membrane lipid constituents. A previous study by our group demonstrated that phosphatidylethanolamine of H. pylori functions as the cholesterol-binding lipid. It is, however, unclear whether H. pylori is equipped with protein molecules involved in the cholesterol uptake. We, therefore, examined H. pylori proteins that tightly bind to cholesterol. As a consequence, H. pylori catalase (KatA) turned out to be a candidate of the cholesterol uptake-associated protein. In addition, an H. pylori mutant strain that expresses KatA protein lacking catalase activity was significantly lower in total cholesterol contents than the wild-type H. pylori strain. The putative amino acid sequence of KatA found out to contain a number of the cholesterol recognition/interaction amino acid consensus sequence domains (CRAC and CARC domains). These results suggest that H. pylori KatA with normal folding conformation acts as the cholesterol-binding or -storage protein.
Collapse
Affiliation(s)
- Kouichi Hosoda
- Nikon Cell Innovation Co., Ltd., 2-4-10, Shinsuna, Koto-ku, Tokyo 136-0075, Japan
| | - Kiyofumi Wanibuchi
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa 245-0066, Japan
| | - Avarzed Amgalanbaatar
- Department of Graduate Education, Graduate School, Mongolian National University of Medical Sciences, 14210, Zoing Street, Sukhbaatar District, Ulaanbaatar 14210, Mongolia
| | - Mitsuru Shoji
- Faculty of Pharmaceutical Sciences, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa 245-0066, Japan
| | - Shunji Hayashi
- Department of Microbiology, Kitasato University School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara-shi, Kanagawa 252-0374, Japan
| | - Hirofumi Shimomura
- Public Health Center of Uki, Kumamoto Prefecture Office, 400-1, Kugu, Matsubase-machi, Uki-shi, Kumamoto 869-0532, Japan.
| |
Collapse
|
18
|
Shimamura M, Kamijo SI, Illarionov P. C-type lectin Mincle-dependent and -independent activation of invariant NKT cells by exposure to Helicobacter pylori α-cholesteryl glucosides. FEBS J 2023; 290:134-147. [PMID: 35920835 DOI: 10.1111/febs.16588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/01/2022] [Accepted: 08/03/2022] [Indexed: 01/14/2023]
Abstract
Helicobacter pylori extracts cholesterol from the host and converts it to its glycosides. We found that cholesteryl 6'-O-acyl α-glucoside (ChAcαG) produced by H. pylori is recognised by both invariant Vα14+ NKT (iNKT) cells and a C-type lectin receptor Mincle (Clec4e). However, it is unclear how these duplicated recognitions cooperate and contribute to host defence against H. pylori. Among T cell populations in the liver, iNKT cells predominantly expressed the T cell activation marker CD69 just after stimulation with ChAcαG. The production of IFN-γ and IL-4 was strictly dependent on both CD1d and Jα18 expressions, indicating the necessity of iNKT cell activation for the initiation of immune responses. Production of IFN-γ by iNKT cells was markedly reduced by the Mincle deficiency on antigen-presenting cells (APCs), while IL-4 production was not significantly influenced. IL-2 production by iNKT cell hybridomas was also diminished by the Mincle deficiency upon stimulation with APCs previously loaded with ChAcαG. Here, the immune responses of iNKT cell hybridomas stimulated with wild-type APCs were reduced by the addition of anti-IL-12 blocking antibody to the level stimulated with Mincle-deficient APCs. Collectively, these results suggest that iNKT cells can be activated with the cholesteryl glycosides via a Mincle-dependent, IL-12 signal-dependent pathway and a Mincle-independent, invariant TCR signal-dominant pathway. iNKT cells activated via the Mincle-dependent pathway produce IFN-γ-dominant cytokines; hence, they may contribute to enhancing proinflammatory responses against H. pylori infection.
Collapse
Affiliation(s)
- Michio Shimamura
- Tsukuba Research Center for Interdisciplinary Materials Science, University of Tsukuba, Japan.,Mitsubishi Kagaku Institute of Life Sciences, Tokyo, Japan.,School of Science and Technology, Meiji University, Kawasaki, Japan
| | - Shin-Ichi Kamijo
- Mitsubishi Kagaku Institute of Life Sciences, Tokyo, Japan.,LifeWill Corporation, Tokyo, Japan
| | | |
Collapse
|
19
|
Fuchs S, Gong R, Gerhard M, Mejías-Luque R. Immune Biology and Persistence of Helicobacter pylori in Gastric Diseases. Curr Top Microbiol Immunol 2023; 444:83-115. [PMID: 38231216 DOI: 10.1007/978-3-031-47331-9_4] [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] [Indexed: 01/18/2024]
Abstract
Helicobacter pylori is a prevalent pathogen, which affects more than 40% of the global population. It colonizes the human stomach and persists in its host for several decades or even a lifetime, if left untreated. The persistent infection has been linked to various gastric diseases, including gastritis, peptic ulcers, and an increased risk for gastric cancer. H. pylori infection triggers a strong immune response directed against the bacterium associated with the infiltration of innate phagocytotic immune cells and the induction of a Th1/Th17 response. Even though certain immune cells seem to be capable of controlling the infection, the host is unable to eliminate the bacteria as H. pylori has developed remarkable immune evasion strategies. The bacterium avoids its killing through innate recognition mechanisms and manipulates gastric epithelial cells and immune cells to support its persistence. This chapter focuses on the innate and adaptive immune response induced by H. pylori infection, and immune evasion strategies employed by the bacterium to enable persistent infection.
Collapse
Affiliation(s)
- Sonja Fuchs
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Ruolan Gong
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany.
| |
Collapse
|
20
|
Helicobacter pylori shows tropism to gastric differentiated pit cells dependent on urea chemotaxis. Nat Commun 2022; 13:5878. [PMID: 36198679 PMCID: PMC9535007 DOI: 10.1038/s41467-022-33165-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/06/2022] [Indexed: 11/09/2022] Open
Abstract
The human gastric epithelium forms highly organized gland structures with different subtypes of cells. The carcinogenic bacterium Helicobacter pylori can attach to gastric cells and subsequently translocate its virulence factor CagA, but the possible host cell tropism of H. pylori is currently unknown. Here, we report that H. pylori preferentially attaches to differentiated cells in the pit region of gastric units. Single-cell RNA-seq shows that organoid-derived monolayers recapitulate the pit region, while organoids capture the gland region of the gastric units. Using these models, we show that H. pylori preferentially attaches to highly differentiated pit cells, marked by high levels of GKN1, GKN2 and PSCA. Directed differentiation of host cells enable enrichment of the target cell population and confirm H. pylori preferential attachment and CagA translocation into these cells. Attachment is independent of MUC5AC or PSCA expression, and instead relies on bacterial TlpB-dependent chemotaxis towards host cell-released urea, which scales with host cell size. The carcinogenic bacterium Helicobacter pylori infects gastric cells. Here, the authors show that H. pylori preferentially infects differentiated cells in the pit region of gastric units, and this relies on bacterial chemotaxis towards host cell-released urea, which scales with host cell size.
Collapse
|
21
|
Inhibitory and Injury-Protection Effects of O-Glycan on Gastric Epithelial Cells Infected with Helicobacter pylori. Infect Immun 2022; 90:e0039322. [PMID: 36190255 PMCID: PMC9584294 DOI: 10.1128/iai.00393-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Helicobacter pylori (H. pylori) is an important pathogen that can cause gastric cancer. Multiple adhesion molecules mediated H. pylori adherence to cells is the initial step in the infection of host cells. H. pylori cholesterol-α-glucosyltransferase (CGT) recognizes and extracts cholesterol from cell membranes to destroy lipid raft structure, further promotes H. pylori adhesion to gastric epithelial cells. O-Glycan, a substance secreted by the deep gastric mucosa, can competitively inhibit CGT activity and may serve as an important factor to prevent H. pylori colonization in the deep gastric mucosa. However, the inhibitory and injury-protection effects of O-Glycan against H. pylori infection has not been well investigated. In this study, we found that O-Glycan significantly inhibited the relative urease content in the coinfection system. In the presence of O-glycan, the injury of GES-1 cells in H. pylori persistent infection model was attenuated and the cell viability was increased. We use fluorescein isothiocyanate-conjugated cholera toxin subunit B (FITC-CTX-B) to detect lipid rafts on gastric epithelial cells and observed that O-glycan can protect H. pylori from damaging lipid raft structures on cell membranes. In addition, transcriptome data showed that O-glycan treatment significantly reduced the activation of inflammatory cancer transformation pathway caused by H. pylori infection. Our results suggest that O-Glycan is able to inhibit H. pylori persistent infection of gastric epithelial cells, reduce the damage caused by H. pylori, and could serve as a potential medicine to treat patients infected with H. pylori.
Collapse
|
22
|
Negative-High Titer of Helicobacter pylori Antibody and Lipid Profiles. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9984255. [PMID: 36017395 PMCID: PMC9398768 DOI: 10.1155/2022/9984255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022]
Abstract
Background. Helicobacter pylori (HP) is a causative factor for several gastrointestinal diseases. A HP seropositive antibody titer (i.e., ≥10 U/mL), a threshold indicating an HP infection, is known to be associated with changes in lipid metabolism. There is evidence that HP infection can be found in some individuals with HP antibody titer of between 3 and 9.9 U/mL (termed as “negative-high titer”). However, it is unknown about the relationship between HP negative-high titer and lipid metabolism. The present study aimed to quantify the association between HP negative-high antibody titer and lipid profiles. Materials and Methods. We surveyed 2,478 people who underwent a Ningen Dock examination and had serological HP antibody data, from May 2016 to December 2020 at National Center for Global Health and Medicine, Tokyo, Japan. Multiple regression models were used to quantify the association between HP antibody titer and serum lipid levels. Results. The adjusted odds ratio (95% confidence interval [CI]) for dyslipidemia in HP negative-high and positive titer was 1.24 (0.96, 1.79) and 1.36 (1.10, 1.68), respectively, compared with HP negative-low titer;
trend =0.005. The adjusted mean (95% CI) of high-density lipoprotein cholesterol (HDL-C) in HP negative-low, negative-high, and positive titer was 58.78 (57.86–59.71), 55.30 (53.70–56.91), and 53.76 (52.90–54.63) mg/dL, respectively;
trend <0.001. Higher HP antibody titers were also associated with higher ratio of low-density lipoprotein cholesterol (LDL-C) to HDL-C, but not triglycerides, or total cholesterols. Conclusion. The present cross-sectional study suggests that a HP negative-high antibody titer may be associated with dyslipidemia, HDL-C, and LDL-C to HDL-C ratio among Japanese Ningen Dock’s participants.
Collapse
|
23
|
Wizenty J, Müllerke S, Kolesnichenko M, Heuberger J, Lin M, Fischer AS, Mollenkopf HJ, Berger H, Tacke F, Sigal M. Gastric stem cells promote inflammation and gland remodeling in response to Helicobacter pylori via Rspo3-Lgr4 axis. EMBO J 2022; 41:e109996. [PMID: 35767364 PMCID: PMC9251867 DOI: 10.15252/embj.2021109996] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 05/17/2022] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori is a pathogen that colonizes the stomach and causes chronic gastritis. Helicobacter pylori can colonize deep inside gastric glands, triggering increased R‐spondin 3 (Rspo3) signaling. This causes an expansion of the “gland base module,” which consists of self‐renewing stem cells and antimicrobial secretory cells and results in gland hyperplasia. The contribution of Rspo3 receptors Lgr4 and Lgr5 is not well explored. Here, we identified that Lgr4 regulates Lgr5 expression and is required for H. pylori‐induced hyperplasia and inflammation, while Lgr5 alone is not. Using conditional knockout mice, we reveal that R‐spondin signaling via Lgr4 drives proliferation of stem cells and also induces NF‐κB activity in the proliferative stem cells. Upon exposure to H. pylori, the Lgr4‐driven NF‐κB activation is responsible for the expansion of the gland base module and simultaneously enables chemokine expression in stem cells, resulting in gland hyperplasia and neutrophil recruitment. This demonstrates a connection between R‐spondin‐Lgr and NF‐κB signaling that links epithelial stem cell behavior and inflammatory responses to gland‐invading H. pylori.
Collapse
Affiliation(s)
- Jonas Wizenty
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Stefanie Müllerke
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Marina Kolesnichenko
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Heuberger
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Manqiang Lin
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Anne-Sophie Fischer
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Hans-Joachim Mollenkopf
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hilmar Berger
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Sigal
- Division of Gastroenterology and Hepatology, Medical Department, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| |
Collapse
|
24
|
Chen X, Zou Y, Zhang S, Fang P, Li S, Li P, Sun Y, Yuan G, Hu H. Multi-functional vesicles improve Helicobacter pylori eradication by a comprehensive strategy based on complex pathological microenvironment. Acta Pharm Sin B 2022; 12:3498-3512. [PMID: 36176916 PMCID: PMC9513559 DOI: 10.1016/j.apsb.2022.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/06/2022] [Accepted: 04/23/2022] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori (H. pylori), creating a global infection rate over 50%, presents great challenges in clinical therapies due to its complex pathological microenvironment in vivo. To improve the eradication efficacy, herein we fabricated a pharmaceutical vesicle RHL/Cl-Ch-cal where cholesterol-PEG, calcitriol and first-line antibiotic clarithromycin were co-loaded in the rhamnolipid-composed outer lipid layer. RHL/Cl-Ch-cal could quickly penetrate through gastric mucus layer to reach H. pylori infection sites, and then effectively destroyed the architecture of H. pylori biofilms, killed dispersed H. pylori and inhibited the re-adhesion of residual bacteria (called biofilms eradication tetralogy). Moreover, RHL/Cl-Ch-cal activated the host immune response to H. pylori by replenishing cholesterol to repair lipid raft on the cell membrane of host epithelial cells. Finally, RHL/Cl-Ch-cal killed the intracellular H. pylori through recovering the lysosomal acidification and assisting degradation. In experiments, RHL/Cl-Ch-cal demonstrated prominent anti-H. pylori efficacy in the classical H. pylori-infected mice model. Therefore, the study provides a “comprehensive attack” strategy for anti-H. pylori therapies including biofilms eradication tetralogy, immune activation and intracellular bacteria killing.
Collapse
|
25
|
Helicobacter pylori Pathogen-Associated Molecular Patterns: Friends or Foes? Int J Mol Sci 2022; 23:ijms23073531. [PMID: 35408892 PMCID: PMC8998707 DOI: 10.3390/ijms23073531] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023] Open
Abstract
Microbial infections are sensed by the host immune system by recognizing signature molecules called Pathogen-Associated Molecular Patterns—PAMPs. The binding of these biomolecules to innate immune receptors, called Pattern Recognition Receptors (PRRs), alerts the host cell, activating microbicidal and pro-inflammatory responses. The outcome of the inflammatory cascade depends on the subtle balance between the bacterial burn and the host immune response. The role of PRRs is to promote the clearance of the pathogen and to limit the infection by bumping inflammatory response. However, many bacteria, including Helicobacter pylori, evolved to escape PRRs’ recognition through different camouflages in their molecular pattern. This review examines all the different types of H. pylori PAMPs, their roles during the infection, and the mechanisms they evolved to escape the host recognition.
Collapse
|
26
|
Schmidinger B, Petri K, Lettl C, Li H, Namineni S, Ishikawa-Ankerhold H, Jiménez-Soto LF, Haas R. Helicobacter pylori binds human Annexins via Lipopolysaccharide to interfere with Toll-like Receptor 4 signaling. PLoS Pathog 2022; 18:e1010326. [PMID: 35176125 PMCID: PMC8890734 DOI: 10.1371/journal.ppat.1010326] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 03/02/2022] [Accepted: 01/31/2022] [Indexed: 12/19/2022] Open
Abstract
Helicobacter pylori colonizes half of the global population and causes gastritis, peptic ulcer disease or gastric cancer. In this study, we were interested in human annexin (ANX), which comprises a protein family with diverse and partly unknown physiological functions, but with a potential role in microbial infections and possible involvement in gastric cancer. We demonstrate here for the first time that H. pylori is able to specifically bind ANXs. Binding studies with purified H. pylori LPS and specific H. pylori LPS mutant strains indicated binding of ANXA5 to lipid A, which was dependent on the lipid A phosphorylation status. Remarkably, ANXA5 binding almost completely inhibited LPS-mediated Toll-like receptor 4- (TLR4) signaling in a TLR4-specific reporter cell line. Furthermore, the interaction is relevant for gastric colonization, as a mouse-adapted H. pylori increased its ANXA5 binding capacity after gastric passage and its ANXA5 incubation in vitro interfered with TLR4 signaling. Moreover, both ANXA2 and ANXA5 levels were upregulated in H. pylori-infected human gastric tissue, and H. pylori can be found in close association with ANXs in the human stomach. Furthermore, an inhibitory effect of ANXA5 binding for CagA translocation could be confirmed. Taken together, our results highlight an adaptive ability of H. pylori to interact with the host cell factor ANX potentially dampening innate immune recognition. H. pylori is very well adapted to its natural habitat, the human gastric mucosa. For this purpose, the bacterium has evolved a number of highly specific virulence factors, such as the cag-type IV secretion system, vacuolating cytotoxin A (VacA) or secreted gamma-glutamyl transpeptidase. An important function of these bacterial factors is to manipulate the host immune response to enable a chronic H. pylori infection. The present work identifies a new player in this process. Here, we have discovered that H. pylori, as well as several other bacterial species, can bind human annexins (ANX), suggesting a more widespread phenomenon. We show that H. pylori specifically binds ANXA5 via lipid A. The interaction is strictly dependent on calcium and modulated by the phosphorylation status of lipid A. Notably, ANXA5 binding strongly inhibits LPS-mediated Toll-like receptor 4 (TLR4) signal transduction, suggesting that H. pylori exploits ANXs binding to avoid its recognition by this important receptor of the innate immune system. The study thus provides novel molecular and mechanistic insights into a further strategy of H. pylori to successfully evade recognition by the host.
Collapse
Affiliation(s)
- Barbara Schmidinger
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Germany
| | - Kristina Petri
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Germany
| | - Clara Lettl
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Germany
| | - Hong Li
- West China Marshall Research Center for Infectious Diseases, Center of Infectious Diseases, Division of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Sukumar Namineni
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Germany
| | - Hellen Ishikawa-Ankerhold
- Department of Internal Medicine I, Faculty of Medicine, LMU Munich, Germany
- Walter Brendel Centre of Experimental Medicine, University Hospital, LMU Munich, Germany
| | - Luisa Fernanda Jiménez-Soto
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Germany
| | - Rainer Haas
- Chair of Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Germany
- German Center for Infection Research (DZIF), LMU Munich, Germany
- * E-mail:
| |
Collapse
|
27
|
Wang X, Jiang D, Li T, Zhang X, Wang R, Gao S, Yang F, Wang Y, Tian Q, Xie C, Liang J. Association between microbiological risk factors and neurodegenerative disorders: An umbrella review of systematic reviews and meta-analyses. Front Psychiatry 2022; 13:991085. [PMID: 36213914 PMCID: PMC9537612 DOI: 10.3389/fpsyt.2022.991085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/30/2022] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED The role of microbiological factors in the development of neurodegenerative diseases is attracting increasing attention, while the relationship remains debated. This study aimed to comprehensively summarize and evaluate the associations between microbiological factors and the risk of neurodegenerative disorders with an umbrella review. PubMed, Embase, and the Cochrane library were used to search for papers from the earliest to March 2021 for identifying meta-analyses and systematic reviews that examined associations between microbiological factors and neurodegenerative diseases. AMSTAR2 tool was employed to evaluate the methodical quality of systematic reviews and meta-analyses. The effect size and 95% confidence interval (95% CI) were recalculated with a random effect model after the overlap was recognized by the corrected covered area (CCA) method. The heterogeneity of each meta-analysis was measured by the I 2 statistic and 95% prediction interval (95% PI). Additionally, publication bias and the quality of evidence were evaluated for all 37 unique associations. Only 4 associations had above the medium level of evidence, and the rest associations presented a low level of evidence. Among them, helicobacter pylori (HP), infection, and bacteria are associated with Parkinson's disease (PD), and the other one verifies that periodontal disease is a risk factor for all types of dementia. Following the evidence of our study, eradication of HP and aggressive treatment of periodontitis are beneficial for the prevention of PD and dementia, respectively. This umbrella review provides comprehensive quality-grade evidence on the relationship between microbial factors and neurodegenerative disease. Regardless of much evidence linking microbial factors to neurodegenerative diseases, these associations are not necessarily causal, and the evidence level is generally low. Thus, more effective studies are required. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/PROSPERO/#searchadvanced, PROSPERO, identifier: CRD42021239512.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing, China
| | - Deming Jiang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tianxiong Li
- Surgery Centre of Diabetes Mellitus, Peking University Ninth School of Clinical Medicine (Beijing Shijitan Hospital, Capital Medical University), Beijing, China
| | - Xiao Zhang
- Department of Neurology, Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing, China
| | - Ran Wang
- Department of Neurology, Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing, China
| | - Song Gao
- Department of Neurology, Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing, China
| | - Fengyi Yang
- Department of Neurology, Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing, China
| | - Yan Wang
- Department of Neurology, Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing, China
| | - Qi Tian
- Department of Neurology, Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing, China
| | - Chunrong Xie
- Department of Neurology, Beijing Huairou Hospital of Traditional Chinese Medicine, Beijing, China
- Chunrong Xie
| | - Jinghong Liang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Jinghong Liang
| |
Collapse
|
28
|
Lim JH, Shin CM, Han K, Yoo J, Jin EH, Choi YJ, Lee DH. Nationwide cohort study: cholesterol level is inversely related with the risk of gastric cancer among postmenopausal women. Gastric Cancer 2022; 25:11-21. [PMID: 34468870 DOI: 10.1007/s10120-021-01241-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent studies showed inverse relationship between hypercholesterolemia and the risk of gastric cancer, especially among male. However evidence among female is inconsistent. We aimed to investigate the relationship between cholesterol level and the risk of gastric cancer among female according to menopausal status. METHODS We analyzed the data from a population-based prospective cohort of female ≥ 30 years old who underwent cancer screening and general health screening provided by the Korean National Health Insurance Corporation in 2009. Under quartile stratification of the level of cholesterol components, we calculated the hazard ratio (HR) for gastric cancer incidence until 2018 for each level group according to the menopausal status at 2009. RESULTS Among total 2,722,614 individuals, 17,649 gastric cancer cases developed after mean 8.26 years of follow-up (premenopausal 3746/1180666; postmenopausal 13,903/1541948). Total cholesterol, high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) showed inverse relationship with the risk of gastric cancer among postmenopausal women (adjusted HR (95% confidence interval) for the highest quartile vs. lowest quartile and p-for-trend: 0.88 (0.84-0.92) and < 0.001 for total cholesterol; 0.89 (0.85-0.92) and < 0.001 for HDL-C; 0.92 (0.89-0.97) and 0.001 for LDL-C), whereas none showed statistically significant risk relationship among premenopausal women. Triglyceride was not independently related with gastric cancer risk among both pre- and postmenopausal women. CONCLUSIONS Cholesterol levels, including total cholesterol, HDL-C, and LDL-C, are inversely related with the risk of gastric cancer among postmenopausal women, but not among premenopausal women.
Collapse
Affiliation(s)
- Joo Hyun Lim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, South Korea
| | - Cheol Min Shin
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea. .,Department of Internal Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, South Korea
| | - Juhwan Yoo
- Department of Biomedicine and Health Science, The Catholic University of Korea, Seoul, Korea
| | - Eun Hyo Jin
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, South Korea
| | - Yoon Jin Choi
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Ho Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| |
Collapse
|
29
|
Macrophages in Microbial Pathogenesis: Commonalities of Defense Evasion Mechanisms. Infect Immun 2021; 90:e0029121. [PMID: 34780281 DOI: 10.1128/iai.00291-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Macrophages are key arsenals of the immune system against invaders. After compartmental isolation of a pathogen in phagosomes, the host immune response attempts to neutralize the pathogen. However, pathogens possess the ability to subvert these assaults and can also convert macrophages into their replicative niche. The multiple host defense evasion mechanisms employed by these pathogens like phagosome maturation arrest, molecular mimicry through secretory antigens, interference with host signaling, active radical neutralization, inhibition of phagosome acidification, alteration of programmed cell death and many other mechanisms. Macrophage biology as a part of the host-pathogen interaction has expanded rapidly in the past decade. The present review aims to shed some light upon the macrophage defense evasion strategies employed by infecting pathogens. We have also incorporated recent knowledge in the field of macrophage dynamics during infection and evolutionary perspectives of macrophage dynamics.
Collapse
|
30
|
Muthusamy S, Jan HM, Hsieh MY, Mondal S, Liu WC, Ko YA, Yang WY, Mong KKT, Chen GC, Lin CH. Enhanced enzymatic production of cholesteryl 6'-acylglucoside impairs lysosomal degradation for the intracellular survival of Helicobacter pylori. J Biomed Sci 2021; 28:72. [PMID: 34706729 PMCID: PMC8549234 DOI: 10.1186/s12929-021-00768-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/08/2021] [Indexed: 01/15/2023] Open
Abstract
Background During autophagy defense against invading microbes, certain lipid types are indispensable for generating specialized membrane-bound organelles. The lipid composition of autophagosomes remains obscure, as does the issue of how specific lipids and lipid-associated enzymes participate in autophagosome formation and maturation. Helicobacter pylori is auxotrophic for cholesterol and converts cholesterol to cholesteryl glucoside derivatives, including cholesteryl 6ʹ-O-acyl-α-d-glucoside (CAG). We investigated how CAG and its biosynthetic acyltransferase assist H. pylori to escape host-cell autophagy. Methods We applied a metabolite-tagging method to obtain fluorophore-containing cholesteryl glucosides that were utilized to understand their intracellular locations. H. pylori 26695 and a cholesteryl glucosyltransferase (CGT)-deletion mutant (ΔCGT) were used as the standard strain and the negative control that contains no cholesterol-derived metabolites, respectively. Bacterial internalization and several autophagy-related assays were conducted to unravel the possible mechanism that H. pylori develops to hijack the host-cell autophagy response. Subcellular fractions of H. pylori-infected AGS cells were obtained and measured for the acyltransferase activity. Results The imaging studies of fluorophore-labeled cholesteryl glucosides pinpointed their intracellular localization in AGS cells. The result indicated that CAG enhances the internalization of H. pylori in AGS cells. Particularly, CAG, instead of CG and CPG, is able to augment the autophagy response induced by H. pylori. How CAG participates in the autophagy process is multifaceted. CAG was found to intervene in the degradation of autophagosomes and reduce lysosomal biogenesis, supporting the idea that intracellular H. pylori is harbored by autophago-lysosomes in favor of the bacterial survival. Furthermore, we performed the enzyme activity assay of subcellular fractions of H. pylori-infected AGS cells. The analysis showed that the acyltransferase is mainly distributed in autophago-lysosomal compartments. Conclusions Our results support the idea that the acyltransferase is mainly distributed in the subcellular compartment consisting of autophagosomes, late endosomes, and lysosomes, in which the acidic environment is beneficial for the maximal acyltransferase activity. The resulting elevated level of CAG can facilitate bacterial internalization, interfere with the autophagy flux, and causes reduced lysosomal biogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-021-00768-w.
Collapse
Affiliation(s)
- Sasikala Muthusamy
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung-Hsing University and Academia Sinica, Taipei, 11529, Taiwan.,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227, Taiwan.,Biotechnology Center, National Chung-Hsing University, Taichung, 40227, Taiwan
| | - Hau-Ming Jan
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Ming-Yen Hsieh
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Soumik Mondal
- Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan
| | - Wen-Chun Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-An Ko
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Wei-Yuan Yang
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Kwok-Kong Tony Mong
- Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan
| | - Guang-Chao Chen
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan. .,Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung-Hsing University and Academia Sinica, Taipei, 11529, Taiwan. .,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227, Taiwan. .,Biotechnology Center, National Chung-Hsing University, Taichung, 40227, Taiwan. .,Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan. .,Department of Chemistry, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
31
|
Hsu CY, Yeh JY, Chen CY, Wu HY, Chiang MH, Wu CL, Lin HJ, Chiu CH, Lai CH. Helicobacter pylori cholesterol-α-glucosyltransferase manipulates cholesterol for bacterial adherence to gastric epithelial cells. Virulence 2021; 12:2341-2351. [PMID: 34506250 PMCID: PMC8437457 DOI: 10.1080/21505594.2021.1969171] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori infection is associated with several gastrointestinal diseases, including gastritis, peptic ulcers, and gastric cancer. Infection of cells with H. pylori is dependent on lipid rafts, which are cholesterol-rich microdomains located in the cell membrane. H. pylori cholesterol-α-glucosyltransferase (CGT) catalyzes the conversion of membrane cholesterol to cholesteryl glucosides, which can be incorporated into the bacterial cell wall, facilitating evasion from immune defense and colonization in the host. However, the detailed mechanisms underlying this process remain to be explored. In this study, we discovered for the first time that H. pylori CGT could promote adherence to gastric epithelial cells in a cholesterol-dependent manner. Externalization of cell membrane phosphatidylserine (PS) is crucial for enhancement of binding of H. pylori to cells by CGT and for cytotoxin-associated gene A (CagA)-induced pathogenesis. Furthermore, exogenous cholesterol interferes with the actions of H. pylori CGT to catalyze cellular cholesterol, which impedes bacterial binding to cells and attenuates subsequent inflammation, indicating that the initial attachment of H. pylori to cells is closely dependent on host cholesterol. These results provide evidence that CGT contributes to H. pylori infectivity and it may serve as a key target for the treatment of H. pylori-associated diseases.
Collapse
Affiliation(s)
- Chung-Yao Hsu
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jia-Yin Yeh
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Ya Chen
- Department of Laboratory Medicine, Taichung Veterans General Hospital Chiayi Branch, Chiayi, Taiwan
| | - Hui-Yu Wu
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Meng-Hsuan Chiang
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chia-Lin Wu
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hwai-Jeng Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang-Ho Hospital, New Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Hsun Chiu
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Ho Lai
- Graduate Institute of Biomedical Sciences, Department of Microbiology and Immunology, Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
32
|
Nagata M, Toyonaga K, Ishikawa E, Haji S, Okahashi N, Takahashi M, Izumi Y, Imamura A, Takato K, Ishida H, Nagai S, Illarionov P, Stocker BL, Timmer MSM, Smith DGM, Williams SJ, Bamba T, Miyamoto T, Arita M, Appelmelk BJ, Yamasaki S. Helicobacter pylori metabolites exacerbate gastritis through C-type lectin receptors. J Exp Med 2021; 218:152132. [PMID: 32991669 PMCID: PMC7527975 DOI: 10.1084/jem.20200815] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/17/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori causes gastritis, which has been attributed to the development of H. pylori-specific T cells during infection. However, the mechanism underlying innate immune detection leading to the priming of T cells is not fully understood, as H. pylori evades TLR detection. Here, we report that H. pylori metabolites modified from host cholesterol exacerbate gastritis through the interaction with C-type lectin receptors. Cholesteryl acyl α-glucoside (αCAG) and cholesteryl phosphatidyl α-glucoside (αCPG) were identified as noncanonical ligands for Mincle (Clec4e) and DCAR (Clec4b1). During chronic infection, H. pylori-specific T cell responses and gastritis were ameliorated in Mincle-deficient mice, although bacterial burdens remained unchanged. Furthermore, a mutant H. pylori strain lacking αCAG and αCPG exhibited an impaired ability to cause gastritis. Thus H. pylori-specific modification of host cholesterol plays a pathophysiological role that exacerbates gastric inflammation by triggering C-type lectin receptors.
Collapse
Affiliation(s)
- Masahiro Nagata
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kenji Toyonaga
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Shojiro Haji
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Nobuyuki Okahashi
- Department of Bioinformatics Engineering, Graduate School of Information Science and Technology, Osaka University, Suita, Osaka, Japan.,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Akihiro Imamura
- Department of Applied Bioorganic Chemistry, Gifu University, Gifu, Gifu, Japan
| | - Koichi Takato
- Department of Applied Bioorganic Chemistry, Gifu University, Gifu, Gifu, Japan
| | - Hideharu Ishida
- Department of Applied Bioorganic Chemistry, Gifu University, Gifu, Gifu, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu, Gifu, Japan
| | - Shigenori Nagai
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Petr Illarionov
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Bridget L Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand.,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Mattie S M Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand.,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Dylan G M Smith
- School of Chemistry and Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Spencer J Williams
- School of Chemistry and Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Tomofumi Miyamoto
- Department of Natural Products Chemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa, Japan.,Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Ben J Appelmelk
- Molecular Microbiology/Medical Microbiology and Infection Control, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Division of Molecular Design, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan.,Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| |
Collapse
|
33
|
Lin TY, Lan WH, Chiu YF, Feng CL, Chiu CH, Kuo CJ, Lai CH. Statins' Regulation of the Virulence Factors of Helicobacter pylori and the Production of ROS May Inhibit the Development of Gastric Cancer. Antioxidants (Basel) 2021; 10:1293. [PMID: 34439541 PMCID: PMC8389206 DOI: 10.3390/antiox10081293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
Conventionally, statins are used to treat high cholesterol levels. They exhibit pleiotropic effects, such as the prevention of cardiovascular disease and decreased cancer mortality. Gastric cancer (GC) is one of the most common cancers, ranking as the third leading global cause of cancer-related deaths, and is mainly attributed to chronic Helicobacter pylori infection. During their co-evolution with hosts, H. pylori has developed the ability to use the cellular components of the host to evade the immune system and multiply in intracellular niches. Certain H. pylori virulence factors, including cytotoxin-associated gene A (CagA), vacuolating cytotoxin A (VacA), and cholesterol-α-glucosyltransferase (CGT), have been shown to exploit host cholesterol during pathogenesis. Therefore, using statins to antagonize cholesterol synthesis might prove to be an ideal strategy for reducing the occurrence of H. pylori-related GC. This review discusses the current understanding of the interplay of H. pylori virulence factors with cholesterol and reactive oxygen species (ROS) production, which may prove to be novel therapeutic targets for the development of effective treatment strategies against H. pylori-associated GC. We also summarize the findings of several clinical studies on the association between statin therapy and the development of GC, especially in terms of cancer risk and mortality.
Collapse
Affiliation(s)
- Ting-Yu Lin
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (T.-Y.L.); (W.-H.L.); (Y.-F.C.); (C.-H.C.)
- Research Center for Emerging Viral, Infections Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Wen-Hsi Lan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (T.-Y.L.); (W.-H.L.); (Y.-F.C.); (C.-H.C.)
- Research Center for Emerging Viral, Infections Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ya-Fang Chiu
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (T.-Y.L.); (W.-H.L.); (Y.-F.C.); (C.-H.C.)
- Research Center for Emerging Viral, Infections Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Medical Laboratory, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Chun-Lung Feng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hsinchu Hospital, Hsinchu 30272, Taiwan;
- Department of Internal Medicine, Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung 40447, Taiwan
| | - Cheng-Hsun Chiu
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (T.-Y.L.); (W.-H.L.); (Y.-F.C.); (C.-H.C.)
- Research Center for Emerging Viral, Infections Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Chia-Jung Kuo
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (T.-Y.L.); (W.-H.L.); (Y.-F.C.); (C.-H.C.)
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Chih-Ho Lai
- Research Center for Emerging Viral, Infections Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Internal Medicine, Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung 40447, Taiwan
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
- Department of Nursing, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
34
|
Timmer MSM, Teunissen TJ, Kodar K, Foster AJ, Yamasaki S, Stocker BL. Cholesteryl glucosides signal through the carbohydrate recognition domain of the macrophage inducible C-type lectin (mincle). Org Biomol Chem 2021; 19:2198-2202. [PMID: 33625427 DOI: 10.1039/d0ob02342f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cholesteryl α-d-glucosides (αGCs) are unique metabolic products of the cancer-causing human pathogen Helicobacter pylori. Via signalling through the Macrophage inducible C-type lectin (Mincle) and the induction of a pro-inflammatory response, they are thought to play a role in the development of gastric atrophy. Herein, we prepared the first library of steryl d-glucosides and determined that they preferentially signal through the carbohydrate recognition domain of human Mincle, rather than the amino acid consensus motif. Lipidated steryl d-glucosides exhibited enhanced Mincle agonist activity, with C18 cholesteryl 6-O-acyl-α-d-glucoside (2c) being the most potent activator of human monocytes. Despite exhibiting strong Mincle signalling, sito- (5b) and stigmasterol glycosides (6b) led to a poor inflammatory response in primary cells, suggesting that Mincle is a potential therapeutic target for preventing H. pylori-mediated inflammation and cancer.
Collapse
Affiliation(s)
- Mattie S M Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| | - Thomas J Teunissen
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| | - Kristel Kodar
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| | - Amy J Foster
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan and Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan and Division of Molecular Design, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan and Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| | - Bridget L Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| |
Collapse
|
35
|
Hove PR, Magunda F, de Mello Marques MA, Islam MN, Harton MR, Jackson M, Belisle JT. Identification and functional analysis of a galactosyltransferase capable of cholesterol glycolipid formation in the Lyme disease spirochete Borrelia burgdorferi. PLoS One 2021; 16:e0252214. [PMID: 34061884 PMCID: PMC8168883 DOI: 10.1371/journal.pone.0252214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/11/2021] [Indexed: 01/22/2023] Open
Abstract
Borrelia burgdorferi (Bb), the etiological agent of Lyme disease, produces a series of simple glycolipids where diacylglycerol and cholesterol serve as the precursor. The cholesterol-based glycolipids, cholesteryl 6-O-acyl-β-D-galactopyranoside (ACGal) and cholesteryl-β-D-galactopyranoside (CGal) are immunogenic and proposed to contribute to the pathogenesis of Lyme disease. Detailed studies of CGal and ACGal in Bb have been hampered by a lack of knowledge of their underlying biosynthetic processes. The genome of Bb encodes four putative glycosyltransferases, and only one of these, BB0572, was predicted to be an inverting family 2 glycosyltransferase (GT2 enzyme) capable of using UDP-galactose as a substrate and forming a β-glycosidic bond. Comparison of the 42 kDa BB0572 amino acid sequence from Bb with other Borrelia spp demonstrates that this protein is highly conserved. To establish BB0572 as the galactosyltransferase capable of cholesterol glycolipid formation in Bb, the protein was produced as a recombinant product in Escherichia coli and tested in a cell-free assay with 14C-cholesterol and UDP-galactose as the substrates. This experiment resulted in a radiolabeled lipid that migrated with the cholesterol glycolipid standard of CGal when evaluated by thin layer chromatography. Additionally, mutation in the predicted active site of BB0572 resulted in a recombinant protein that was unable to catalyze the formation of the cholesterol glycolipid. These data characterize BB0572 as a putative cholesterol galactosyltransferase. This provides the first step in understanding how Bb cholesterol glycolipids are formed and will allow investigations into their involvement in pathogen transmission and disease development.
Collapse
Affiliation(s)
- Petronella R. Hove
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States of America
| | - Forgivemore Magunda
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States of America
| | - Maria Angela de Mello Marques
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States of America
| | - M. Nurul Islam
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States of America
| | - Marisa R. Harton
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States of America
| | - Mary Jackson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States of America
| | - John T. Belisle
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States of America
| |
Collapse
|
36
|
Imai S, Ooki T, Murata-Kamiya N, Komura D, Tahmina K, Wu W, Takahashi-Kanemitsu A, Knight CT, Kunita A, Suzuki N, Del Valle AA, Tsuboi M, Hata M, Hayakawa Y, Ohnishi N, Ueda K, Fukayama M, Ushiku T, Ishikawa S, Hatakeyama M. Helicobacter pylori CagA elicits BRCAness to induce genome instability that may underlie bacterial gastric carcinogenesis. Cell Host Microbe 2021; 29:941-958.e10. [PMID: 33989515 DOI: 10.1016/j.chom.2021.04.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/17/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Infection with CagA-producing Helicobacter pylori plays a causative role in the development of gastric cancer. Upon delivery into gastric epithelial cells, CagA deregulates prooncogenic phosphatase SHP2 while inhibiting polarity-regulating kinase PAR1b through complex formation. Here, we show that CagA/PAR1b interaction subverts nuclear translocation of BRCA1 by inhibiting PAR1b-mediated BRCA1 phosphorylation. It hereby induces BRCAness that promotes DNA double-strand breaks (DSBs) while disabling error-free homologous recombination-mediated DNA repair. The CagA/PAR1b interaction also stimulates Hippo signaling that circumvents apoptosis of DNA-damaged cells, giving cells time to repair DSBs through error-prone mechanisms. The DSB-activated p53-p21Cip1 axis inhibits proliferation of CagA-delivered cells, but the inhibition can be overcome by p53 inactivation. Indeed, sequential pulses of CagA in TP53-mutant cells drove somatic mutation with BRCAness-associated genetic signatures. Expansion of CagA-delivered cells with BRCAness-mediated genome instability, from which CagA-independent cancer-predisposing cells arise, provides a plausible "hit-and-run mechanism" of H. pylori CagA for gastric carcinogenesis.
Collapse
Affiliation(s)
- Satoshi Imai
- Department of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Takuya Ooki
- Department of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Naoko Murata-Kamiya
- Department of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan.
| | - Daisuke Komura
- Department of Preventive Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Kamrunnesa Tahmina
- Department of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Weida Wu
- Department of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | | | - Christopher Takaya Knight
- Department of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Akiko Kunita
- Department of Pathology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Nobumi Suzuki
- Department of Gastroenterology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Adriana A Del Valle
- Department of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Mayo Tsuboi
- Department of Gastroenterology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Masahiro Hata
- Department of Gastroenterology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Naomi Ohnishi
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Koji Ueda
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Masanori Hatakeyama
- Department of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
37
|
Maia A, Gu Z, Koch A, Berdiel‐Acer M, Will R, Schlesner M, Wiemann S. IFNβ1 secreted by breast cancer cells undergoing chemotherapy reprograms stromal fibroblasts to support tumour growth after treatment. Mol Oncol 2021; 15:1308-1329. [PMID: 33476079 PMCID: PMC8096792 DOI: 10.1002/1878-0261.12905] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/17/2022] Open
Abstract
Chemotherapy (CTX) remains the standard of care for most aggressive tumours, including breast cancer (BC). In BC chemotherapeutic regimens, the maximum tolerated dose of cytotoxic drugs is administered at regular intervals, and cancer cells can re-grow or adapt during the resting periods between cycles. The impact of the tumour microenvironment on the fate of cancer cells after CTX remains poorly understood. Here, we show that paracrine signalling from CTX-treated cancer cells to stromal fibroblasts can drive cancer cell recovery after cytotoxic drug withdrawal. Interferon β1 (IFNβ1) secreted by cancer cells following treatment with high doses of CTX instigates the acquisition of an anti-viral state in stromal fibroblasts. This state is associated with an expression pattern here referred to as interferon signature (IFNS), which encompasses several interferon-stimulated genes (ISGs), including numerous pro-inflammatory cytokine genes. This crosstalk is an important driver of the expansion of BC cells after CTX, and IFNβ1 blockade in tumour cells abrogated their fibroblast-dependent recovery potential. Analysis of human breast carcinomas supported a link between CTX-induced IFNS in tumour stroma and poor response to CTX treatment. First, IFNβ1 expression in human breast carcinomas was found to inversely correlate with recurrence free survival (RFS). Second, using laser capture microdissection data sets, we show a higher expression of IFNS in the stromal tumour compartment compared to the epithelial one and this signature was found to be more prominent in more aggressive subtypes of BC (basal-like), pointing to a pro-tumorigenic role of this signature. Moreover, IFNS was associated with higher recurrence rates and a worse outcome in BC patients. Our study unravels a novel form of paracrine communication between cancer cells and fibroblasts that ultimately results in CTX resistance. Targeting this axis has the potential to improve CTX outcomes in patients with BC.
Collapse
Affiliation(s)
- Ana Maia
- Division of Molecular Genome AnalysisGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Faculty of BiosciencesUniversity of HeidelbergGermany
| | - Zuguang Gu
- Computational OncologyMolecular Diagnostics ProgramNational Center for Tumour Diseases (NCT) and German Cancer Research Center (DKFZ)HeidelbergGermany
- DKFZ‐HIPO (Heidelberg Center for Personalized Oncology)Germany
| | - André Koch
- Department of Women's Health TübingenEberhard‐Karls‐UniversityTübingenGermany
| | - Mireia Berdiel‐Acer
- Division of Molecular Genome AnalysisGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Rainer Will
- Genomics and Proteomics Core FacilityGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Matthias Schlesner
- Bioinformatics and Omics Data AnalyticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Stefan Wiemann
- Division of Molecular Genome AnalysisGerman Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
38
|
Go DM, Lee SH, Lee SH, Woo SH, Kim K, Kim K, Park KS, Park JH, Ha SJ, Kim WH, Choi JH, Kim DY. Programmed Death Ligand 1-Expressing Classical Dendritic Cells MitigateHelicobacter-Induced Gastritis. Cell Mol Gastroenterol Hepatol 2021; 12:715-739. [PMID: 33894424 PMCID: PMC8267570 DOI: 10.1016/j.jcmgh.2021.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Helicobacter pylori has been reported to modulate local immune responses to colonize persistently in gastric mucosa. Although the induced expression of programmed cell death ligand 1 (PD-L1) has been suggested as an immune modulatory mechanism for persistent infection of H pylori, the main immune cells expressing PD-L1 and their functions in Helicobacter-induced gastritis still remain to be elucidated. METHODS The blockades of PD-L1 with antibody or PD-L1-deficient bone marrow transplantation were performed in Helicobacter-infected mice. The main immune cells expressing PD-L1 in Helicobacter-infected stomach were determined by flow cytometry and immunofluorescence staining. Helicobacter felis or H pylori-infected dendritic cell (DC)-deficient mouse models including Flt3-/-, Zbtb46-diphtheria toxin receptor, and BDCA2-diphtheria toxin receptor mice were analyzed for pathologic changes and colonization levels. Finally, the location of PD-L1-expressing DCs and the correlation with H pylori infection were analyzed in human gastric tissues using multiplexed immunohistochemistry. RESULTS Genetic or antibody-mediated blockade of PD-L1 aggravated Helicobacter-induced gastritis with mucosal metaplasia. Gastric classical DCs expressed considerably higher levels of PD-L1 than other immune cells and co-localized with T cells in gastritis lesions from Helicobacter-infected mice and human beings. H felis- or H pylori-infected Flt3-/- or classical DC-depleted mice showed aggravated gastritis with severe T-cell and neutrophil accumulation with low bacterial loads compared with that in control mice. Finally, PD-L1-expressing DCs were co-localized with T cells and showed a positive correlation with H pylori infection in human subjects. CONCLUSIONS The PD-1/PD-L1 pathway may be responsible for the immune modulatory function of gastric DCs that protects the gastric mucosa from Helicobacter-induced inflammation, but allows persistent Helicobacter colonization.
Collapse
Affiliation(s)
- Du-Min Go
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Seung Hyun Lee
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Su-Hyung Lee
- Division of Cancer Biology, Research Institute of National Cancer Center, Gyeonggi-do, Republic of Korea
| | - Sang-Ho Woo
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kibyeong Kim
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Kyeongdae Kim
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Kyu Seong Park
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Woo Ho Kim
- Department of Pathology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Sciences, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea.
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Ammendolia DA, Bement WM, Brumell JH. Plasma membrane integrity: implications for health and disease. BMC Biol 2021; 19:71. [PMID: 33849525 PMCID: PMC8042475 DOI: 10.1186/s12915-021-00972-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Plasma membrane integrity is essential for cellular homeostasis. In vivo, cells experience plasma membrane damage from a multitude of stressors in the extra- and intra-cellular environment. To avoid lethal consequences, cells are equipped with repair pathways to restore membrane integrity. Here, we assess plasma membrane damage and repair from a whole-body perspective. We highlight the role of tissue-specific stressors in health and disease and examine membrane repair pathways across diverse cell types. Furthermore, we outline the impact of genetic and environmental factors on plasma membrane integrity and how these contribute to disease pathogenesis in different tissues.
Collapse
Affiliation(s)
- Dustin A Ammendolia
- Cell Biology Program, Hospital for Sick Children, 686 Bay Street PGCRL, Toronto, ON, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - William M Bement
- Center for Quantitative Cell Imaging and Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - John H Brumell
- Cell Biology Program, Hospital for Sick Children, 686 Bay Street PGCRL, Toronto, ON, M5G 0A4, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A1, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A1, Canada. .,SickKids IBD Centre, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
40
|
A short review, effect of dimethyl-β-cyclodextrin on the interaction between Helicobacter pylori and steroidal compounds. Heliyon 2021; 7:e06767. [PMID: 33912723 PMCID: PMC8065201 DOI: 10.1016/j.heliyon.2021.e06767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/08/2021] [Accepted: 04/08/2021] [Indexed: 01/22/2023] Open
Abstract
The 2,6-di-O-methyl-β-cyclodextrin (dMβCD) is an amphiphilic annular compound consisting of seven dimethyl-glucose molecules. This compound is well known as a solubilizer of lipophilic compounds. Especially, dMβCD extracts cholesterol from the plasma membrane of mammalian cells and releases the cholesterol to the aqueous solution. The experimental use of dMβCD, therefore, serves to investigate the role of cholesterol in the mammalian cell membrane. It is, however, unclear as to how dMβCD extracts cholesterol incorporated into the glycerophospholipid biomembrane. Meanwhile, dMβCD acts as a beneficial compound for Helicobacter pylori and is used as the standard component for supporting the growth of this bacterium in the serum-free culture. However, the detailed mechanism of dMβCD for supporting the growth of H. pylori is still to be clarified. H. pylori is a Gram-negative microaerophilic bacillus recognized as a pathogen concerned with gastrointestinal diseases in human. Previous studies by our group have successfully obtained the H. pylori strains culturable without dMβCD and demonstrated the distinct effects of dMβCD on the interaction between H. pylori and exogenous steroidal compounds. For instance, dMβCD promotes and inhibits the absorption of cholesterol and several steroidal compounds respectively into the biomembranes of H. pylori. In this study we summarized behaviors of dMβCD toward steroidal compounds relevant to H. pylori.
Collapse
|
41
|
Qaria MA, Qumar S, Sepe LP, Ahmed N. Cholesterol glucosylation-based survival strategy in Helicobacter pylori. Helicobacter 2021; 26:e12777. [PMID: 33368895 DOI: 10.1111/hel.12777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022]
Abstract
Helicobacter pylori is a major chronic health problem, infecting more than half of the population worldwide. H. pylori infection is linked with various clinical complications ranging from gastritis to gastric cancer. The resolution of gastritis and peptic ulcer appears to be linked with the eradication of H. pylori. However, resistance to antibiotics and eradication failure rates are reaching alarmingly high levels. This calls for urgent action in finding alternate methods for H. pylori eradication. Here, we discuss the recently identified mechanism of H. pylori known as cholesterol glucosylation, mediated by the enzyme cholesterol-α-glucosyltransferase, encoded by the gene cgt. Cholesterol glucosylation serves several functions that include promoting immune evasion, enhancing antibiotic resistance, maintaining the native helical morphology, and supporting functions of prominent virulence factors such as CagA and VacA. Consequently, strategies aiming at inhibition of the cholesterol glucosylation process have the potential to attenuate the potency of H. pylori infection and abrogate H. pylori immune evasion capabilities. Knockout of H. pylori cgt results in unsuccessful colonization and elimination by the host immune responses. Moreover, blocking cholesterol glucosylation can reverse antibiotic susceptibility in H. pylori. In this work, we review the main roles of cholesterol glucosylation in H. pylori and evaluate whether this mechanism can be targeted for the development of alternate methods for eradication of H. pylori infection.
Collapse
Affiliation(s)
- Majjid A Qaria
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| | - Shamsul Qumar
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| | - Ludovico P Sepe
- Department of Biological Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Niyaz Ahmed
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| |
Collapse
|
42
|
Leal YA, Song M, Zabaleta J, Medina-Escobedo G, Caron P, Lopez-Colombo A, Guillemette C, Camargo MC. Circulating Levels of Sex Steroid Hormones and Gastric Cancer. Arch Med Res 2021; 52:660-664. [PMID: 33781580 DOI: 10.1016/j.arcmed.2021.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 02/22/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS Men develop gastric cancer more frequently than women, yet little is known about the mechanisms underlying this sex difference. Sex steroid hormones may influence gastric cancer risk. We therefore assessed whether major circulating adrenal precursors, androgens and estrogens were associated with gastric cancer in a high-risk Mexican population. METHODS Blood samples were collected at time of diagnosis from 50 noncardia gastric cancer patients and 50 histologically confirmed non-atrophic gastritis controls. Serum levels of estradiol, testosterone and dehydroepiandrosterone (DHEA) measured with a validated mass spectrometry method were categorized in tertiles as low (T1), middle (T2), and high (T3). Unconditional logistic regression models were used to calculate odds ratios (ORs) and 95% confidence intervals (CI), adjusting for age, sex, and education. RESULTS Levels of DHEA were inversely associated with gastric cancer (p-trend per tertile increase: <0.0001), with adjusted ORs (95% CI) of T2 and T3 (vs. T1) of 0.25 (0.09-0.70) and 0.10 (0.03-0.34), respectively. Levels of estradiol and testosterone were not significantly associated with gastric cancer. CONCLUSIONS Our study provides evidence that higher concentration of circulating DHEA may be associated with lower risk of noncardia gastric cancer. Longitudinal studies are needed to evaluate the temporality of this association and investigate mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Yelda A Leal
- Centro Institucional de Capacitación y Registro de Cáncer, Unidad Médica de Alta Especialidad, Centro Médico Nacional Ignacio García Téllez, Instituto Mexicano de Seguro Social, Mérida, Yucatán, México.
| | - Minkyo Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Jovanny Zabaleta
- Department of Pediatrics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Gilberto Medina-Escobedo
- Departamento de Patología, Unidad Médica de Alta Especialidad, Centro Médico Nacional Ignacio García Téllez, Instituto Mexicano de Seguro Social, Mérida, Yucatán, México
| | - Patrick Caron
- Pharmacogenomics Laboratory, Centre Hospitalier de l'Universite Laval de Quebec, Research Center and Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - Aurelio Lopez-Colombo
- Unidad Médica de Alta Especialidad, Centro Médico Nacional Manuel Ávila Camacho, Instituto Mexicano de Seguro Social, Puebla, Puebla, México
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier de l'Universite Laval de Quebec, Research Center and Faculty of Pharmacy, Laval University, Quebec City, Quebec, Canada
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| |
Collapse
|
43
|
Structure, metabolism and biological functions of steryl glycosides in mammals. Biochem J 2021; 477:4243-4261. [PMID: 33186452 PMCID: PMC7666875 DOI: 10.1042/bcj20200532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/09/2020] [Accepted: 10/21/2020] [Indexed: 12/20/2022]
Abstract
Steryl glycosides (SGs) are sterols glycosylated at their 3β-hydroxy group. They are widely distributed in plants, algae, and fungi, but are relatively rare in bacteria and animals. Glycosylation of sterols, resulting in important components of the cell membrane SGs, alters their biophysical properties and confers resistance against stress by freezing or heat shock to cells. Besides, many biological functions in animals have been suggested from the observations of SG administration. Recently, cholesteryl glucosides synthesized via the transglycosidation by glucocerebrosidases (GBAs) were found in the central nervous system of animals. Identification of patients with congenital mutations in GBA genes or availability of respective animal models will enable investigation of the function of such endogenously synthesized cholesteryl glycosides by genetic approaches. In addition, mechanisms of the host immune responses against pathogenic bacterial SGs have partially been resolved. This review is focused on the biological functions of SGs in mammals taking into consideration their therapeutic applications in the future.
Collapse
|
44
|
Yeh JY, Lin HJ, Kuo CJ, Feng CL, Chou CH, Lin CD, Wu HY, Li CY, Chiu CH, Lai CH. Campylobacter jejuni Cytolethal Distending Toxin C Exploits Lipid Rafts to Mitigate Helicobacter pylori-Induced Pathogenesis. Front Cell Dev Biol 2021; 8:617419. [PMID: 33708766 PMCID: PMC7940356 DOI: 10.3389/fcell.2020.617419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/28/2020] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori infection is associated with several gastrointestinal diseases, including gastritis, peptic ulcer, and gastrointestinal adenocarcinoma. Two major cytotoxins, vacuolating cytotoxin A (VacA) and cytotoxin-associated gene A (CagA), interact closely with lipid rafts, contributing to H. pylori-associated disease progression. The Campylobacter jejuni cytolethal distending toxin consists of three subunits: CdtA, CdtB, and CdtC. Among them, CdtA and CdtC bind to membrane lipid rafts, which is crucial for CdtB entry into cells. In this study, we employed recombinant CdtC (rCdtC) to antagonize the functions of H. pylori cytotoxin in cells. Our results showed that rCdtC alleviates cell vacuolation induced by H. pylori VacA. Furthermore, rCdtC reduces H. pylori CagA translocation, which decreases nuclear factor kappa-B activation and interleukin-8 production, resulting in the mitigation of gastric epithelial cell inflammation. These results reveal that CdtC hijacks cholesterol to compete for H. pylori cytotoxin actions via lipid rafts, ameliorating H. pylori-induced pathogenesis.
Collapse
Affiliation(s)
- Jia-Yin Yeh
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hwai-Jeng Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang-Ho Hospital, New Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Jung Kuo
- Chang Gung Microbiota Therapy Center, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Lung Feng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hsinchu Hospital, Hsinchu, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Huei Chou
- Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Infectious Disease, Department of Otolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Der Lin
- Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Infectious Disease, Department of Otolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Hui-Yu Wu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chen-Yi Li
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
45
|
Baj J, Forma A, Sitarz M, Portincasa P, Garruti G, Krasowska D, Maciejewski R. Helicobacter pylori Virulence Factors-Mechanisms of Bacterial Pathogenicity in the Gastric Microenvironment. Cells 2020; 10:E27. [PMID: 33375694 PMCID: PMC7824444 DOI: 10.3390/cells10010027] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer constitutes one of the most prevalent malignancies in both sexes; it is currently the fourth major cause of cancer-related deaths worldwide. The pathogenesis of gastric cancer is associated with the interaction between genetic and environmental factors, among which infection by Helicobacter pylori (H. pylori) is of major importance. The invasion, survival, colonization, and stimulation of further inflammation within the gastric mucosa are possible due to several evasive mechanisms induced by the virulence factors that are expressed by the bacterium. The knowledge concerning the mechanisms of H. pylori pathogenicity is crucial to ameliorate eradication strategies preventing the possible induction of carcinogenesis. This review highlights the current state of knowledge and the most recent findings regarding H. pylori virulence factors and their relationship with gastric premalignant lesions and further carcinogenesis.
Collapse
Affiliation(s)
- Jacek Baj
- Department of Anatomy, Medical University of Lublin, 20-400 Lublin, Poland;
| | - Alicja Forma
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Monika Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Piero Portincasa
- Clinica Medica “Augusto Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Gabriella Garruti
- Section of Endocrinology, Department of Emergency and Organ Transplantations, University of Bari “Aldo Moro” Medical School, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Danuta Krasowska
- Department of Dermatology, Venerology and Paediatric Dermatology of Medical University of Lublin, 20-081 Lublin, Poland;
| | | |
Collapse
|
46
|
Smith DGM, Ito E, Yamasaki S, Williams SJ. Cholesteryl 6- O-acyl-α-glucosides from diverse Helicobacter spp. signal through the C-type lectin receptor Mincle. Org Biomol Chem 2020; 18:7907-7915. [PMID: 32996960 DOI: 10.1039/d0ob01776k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Helicobacter spp. are Gram-negative bacteria that cause a spectrum of disease in the gut, biliary tree and liver. Many Helicobacter spp. produce a range of cholesteryl α-glucosides that have the potential to act as pathogen associated molecular patterns. We report a highly stereoselective α-glucosylation of cholesterol using 3,4,6-tri-O-acetyl-2-O-benzyl-d-glucopyranosyl N-phenyl-2,2,2-trifluoroacetimidate, which allowed the synthesis of cholesteryl α-glucoside (αCG) and representative Helicobacter spp. cholesteryl 6-O-acyl-α-glucosides (αCAGs; acyl = C12:0, 14:0, C16:0, C18:0, C18:1). All αCAGs, irrespective of the nature of their acyl chain composition, strongly agonised signalling through the C-type lectin receptor Mincle from human and mouse to similar degrees. By contrast, αCG only weakly signalled through human Mincle, and did not signal through mouse Mincle. These results provide a molecular basis for understanding of the immunobiology of non-pylori Helicobacter infections in humans and other animals.
Collapse
Affiliation(s)
- Dylan G M Smith
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia.
| | | | | | | |
Collapse
|
47
|
Pal S, Sarker N, Qaria M, Tandon K, Akhter Y, Ahmed N. Design of an inhibitor of Helicobacter pylori cholesteryl-α-glucoside transferase critical for bacterial colonization. Helicobacter 2020; 25:e12720. [PMID: 32668502 DOI: 10.1111/hel.12720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/23/2020] [Accepted: 06/08/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND Fifty percent of the world's population surves as a host for Helicobacter pylori, gastric cancer causing bacteria, that colonizes the gastric region of digestive tract. It has a remarkable capacity to infect the host stomach for the entire lifetime despite an activated host immune response. METHODS In this study, we have performed the virtual screening analysis of protein-inhibitor binding between the glycosyl transferase enzymes of Helicobacter pylori (CapJ or HP0421) and a corresponding library of inhibitors in the known substrate-binding pockets. We have docked our library of ligands consisting of cholesterol backbone with CapJ protein and identified several ligands' interacting amino acid residues present in active site pocket(s) of the protein. RESULTS In most of the cases, the ligands showed an interaction with the residues of the same pocket of the enzyme. Top three (03) hits were filtered out from the whole data set, which might act as potent inhibitors of the enzyme-substrate reaction. CONCLUSIONS This study describes a new possibility by which colonization of H. pylori can be limited. The reported evidence suggests that comprehensive knowledge and wet laboratory validation of these inhibitors are needed in order to develop them as lead molecules.
Collapse
Affiliation(s)
- Soumiya Pal
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Nishat Sarker
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka
| | - Majjid Qaria
- Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| | - Kshitij Tandon
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Niyaz Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka.,Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| |
Collapse
|
48
|
Bukrinsky MI, Mukhamedova N, Sviridov D. Lipid rafts and pathogens: the art of deception and exploitation. J Lipid Res 2020; 61:601-610. [PMID: 31615838 PMCID: PMC7193957 DOI: 10.1194/jlr.tr119000391] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
Lipid rafts, solid regions of the plasma membrane enriched in cholesterol and glycosphingolipids, are essential parts of a cell. Functionally, lipid rafts present a platform that facilitates interaction of cells with the outside world. However, the unique properties of lipid rafts required to fulfill this function at the same time make them susceptible to exploitation by pathogens. Many steps of pathogen interaction with host cells, and sometimes all steps within the entire lifecycle of various pathogens, rely on host lipid rafts. Such steps as binding of pathogens to the host cells, invasion of intracellular parasites into the cell, the intracellular dwelling of parasites, microbial assembly and exit from the host cell, and microbe transfer from one cell to another all involve lipid rafts. Interaction also includes modification of lipid rafts in host cells, inflicted by pathogens from both inside and outside the cell, through contact or remotely, to advance pathogen replication, to utilize cellular resources, and/or to mitigate immune response. Here, we provide a systematic overview of how and why pathogens interact with and exploit host lipid rafts, as well as the consequences of this interaction for the host, locally and systemically, and for the microbe. We also raise the possibility of modulation of lipid rafts as a therapeutic approach against a variety of infectious agents.
Collapse
Affiliation(s)
- Michael I Bukrinsky
- Department of Microbiology, Immunology, and Tropical Medicine,George Washington University School of Medicine and Health Science, Washington, DC 20037
| | | | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne 3004, Australia. mailto:
| |
Collapse
|
49
|
So AR, Si JM, Lopez D, Pellegrini M. Molecular signatures for inflammation vary across cancer types and correlate significantly with tumor stage, sex and vital status of patients. PLoS One 2020; 15:e0221545. [PMID: 32330128 PMCID: PMC7182171 DOI: 10.1371/journal.pone.0221545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 02/19/2020] [Indexed: 01/02/2023] Open
Abstract
Cancer affects millions of individuals worldwide. One shortcoming of traditional cancer classification systems is that, even for tumors affecting a single organ, there is significant molecular heterogeneity. Precise molecular classification of tumors could be beneficial in personalizing patients’ therapy and predicting prognosis. To this end, here we propose to use molecular signatures to further refine cancer classification. Molecular signatures are collections of genes characterizing particular cell types, tissues or disease. Signatures can be used to interpret expression profiles from heterogeneous samples. Large collections of gene signatures have previously been cataloged in the MSigDB database. We have developed a web-based Signature Visualization Tool (SaVanT) to display signature scores in user-generated expression data. Here we have undertaken a systematic analysis of correlations between inflammatory signatures and cancer samples, to test whether inflammation can differentiate cancer types. Inflammatory response signatures were obtained from MsigDB and SaVanT and a signature score was computed for samples associated with 7 different cancer types. We first identified types of cancers that had high inflammation levels as measured by these signatures. The correlation between signature scores and metadata of these patients (sex, age at initial cancer diagnosis, cancer stage, and vital status) was then computed. We sought to evaluate correlations between inflammation with other clinical parameters and identified four cancer types that had statistically significant association (p-value < 0.05) with at least one clinical characteristic: pancreas adenocarcinoma (PAAD), cholangiocarcinoma (CHOL), kidney chromophobe (KICH), and uveal melanoma (UVM). These results may allow future studies to use these approaches to further refine cancer subtyping and ultimately treatment.
Collapse
Affiliation(s)
- Alexandra Renee So
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| | - Jeong Min Si
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - David Lopez
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
- Gilead Pharmaceuticals, Foster City, California, United States of America
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
50
|
Unique responses of Helicobacter pylori to exogenous hydrophobic compounds. Chem Phys Lipids 2020; 229:104908. [PMID: 32259519 DOI: 10.1016/j.chemphyslip.2020.104908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/14/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori is a pathogen responsible for peptic ulcers and gastric cancers in human. One of the unique biological features of this bacterium is a membrane lipid composition significantly differed from that of typical Gram-negative bacteria. Due to its unique lipid composition, the responses of H. pylori to various exogenous lipophilic compounds significantly differ from the responses of typical Gram-negative bacteria to the same lipophilic compounds. For instance, some steroidal compounds are incorporated into the biomembranes of H. pylori through the intermediation of the myristoyl-phosphatidylethanolamine (PE). In addition, H. pylori shows high susceptibility to bacteriolytic action of lipids such as 3-carbonyl steroids, vitamin D, and indene compounds. These lipids are also considered to interact with myristoyl-PE of H. pylori membranes, and to ultimately confer the bactericidal action to this bacterium. In this study we summarize the lipids concerned with H. pylori and suggest the possibility of the development of chemotherapeutic medicines that act on the membrane lipid component of H. pylori.
Collapse
|